EP1993565A2 - Nutraceutical compositions from microalgae and related methods of production and administration - Google Patents

Nutraceutical compositions from microalgae and related methods of production and administration

Info

Publication number
EP1993565A2
EP1993565A2 EP07808975A EP07808975A EP1993565A2 EP 1993565 A2 EP1993565 A2 EP 1993565A2 EP 07808975 A EP07808975 A EP 07808975A EP 07808975 A EP07808975 A EP 07808975A EP 1993565 A2 EP1993565 A2 EP 1993565A2
Authority
EP
European Patent Office
Prior art keywords
composition
microalgae
cell
cells
polysaccharide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP07808975A
Other languages
German (de)
French (fr)
Inventor
Harrison F. Dillon
Aravind Somanchi
Kamalesh Rao
Peter J.H. Jones
Jonathan Wolfson
Anwar Zaman
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
TerraVia Holdings Inc
Original Assignee
Solazyme Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US11/336,426 external-priority patent/US20070191303A1/en
Priority claimed from US11/337,103 external-priority patent/US20070166266A1/en
Priority claimed from US11/336,431 external-priority patent/US20070166449A1/en
Priority claimed from US11/337,171 external-priority patent/US20070167398A1/en
Priority claimed from US11/336,430 external-priority patent/US20070167397A1/en
Priority claimed from US11/336,428 external-priority patent/US20070167396A1/en
Priority claimed from US11/336,656 external-priority patent/US20070166797A1/en
Application filed by Solazyme Inc filed Critical Solazyme Inc
Publication of EP1993565A2 publication Critical patent/EP1993565A2/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P19/00Preparation of compounds containing saccharide radicals
    • C12P19/04Polysaccharides, i.e. compounds containing more than five saccharide radicals attached to each other by glycosidic bonds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/02Algae
    • A61K36/04Rhodophycota or rhodophyta (red algae), e.g. Porphyra
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/72Cosmetics or similar toiletry preparations characterised by the composition containing organic macromolecular compounds
    • A61K8/73Polysaccharides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • A61Q19/08Anti-ageing preparations
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08BPOLYSACCHARIDES; DERIVATIVES THEREOF
    • C08B37/00Preparation of polysaccharides not provided for in groups C08B1/00 - C08B35/00; Derivatives thereof
    • C08B37/0003General processes for their isolation or fractionation, e.g. purification or extraction from biomass
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08LCOMPOSITIONS OF MACROMOLECULAR COMPOUNDS
    • C08L5/00Compositions of polysaccharides or of their derivatives not provided for in groups C08L1/00 or C08L3/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2800/00Properties of cosmetic compositions or active ingredients thereof or formulation aids used therein and process related aspects
    • A61K2800/74Biological properties of particular ingredients
    • A61K2800/78Enzyme modulators, e.g. Enzyme agonists
    • A61K2800/782Enzyme inhibitors; Enzyme antagonists
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2800/00Properties of cosmetic compositions or active ingredients thereof or formulation aids used therein and process related aspects
    • A61K2800/80Process related aspects concerning the preparation of the cosmetic composition or the storage or application thereof
    • A61K2800/91Injection

Definitions

  • Carbohydrates have the general molecular formula CH 2 O, and thus were once thought to represent "hydrated carbon". However, the arrangement of atoms in carbohydrates has little to do with water molecules.
  • Starch and cellulose are two common carbohydrates. Both are macromolecules with molecular weights in the hundreds of thousands. Both are polymers; that is, each is built from repeating units, monomers, much as a chain is built from its links.
  • Two monosaccharides can be linked together to form a "double" sugar or disaccharide.
  • Three common disaccharides are sucrose, common table sugar (glucose + fructose); lactose, the major sugar in milk (glucose + galactose); and maltose, the product of starch digestion (glucose + glucose).
  • the present invention relates to polysaccharides from microalgae.
  • Representative polysaccharides include those present in the cell wall of microalgae as well as secreted polysaccharides, or exopolysaccharides.
  • the invention includes a variety of compositions containing one or more microalgal polysaccharides as disclosed herein.
  • the compositions include nutraceutical, cosmeceutical, industrial and pharmaceutical compositions which may be used for a variety of indications and uses as described herein.
  • Other compositions include those containing one or more microalgal polysaccharides and a suitable carrier or excipient for topical or oral administration.
  • the invention further relates to methods of producing or preparing microalgal polysaccharides.
  • exogenous sugars are incorporated into the polysaccharides to produce polysaccharides distinct from those present in microalgae that do not incorporate exogenous sugars.
  • the invention also includes methods of trophic conversion and recombinant gene expression in microalgae.
  • recombinant microalgae are prepared to express heterologous gene products, such as mammalian proteins as a non-limiting example, while in other embodiments, the microalgae are modified to produce more of a small molecule already made by microalgae in the absence of genetic modification.
  • the invention relates to methods of using the polysaccharides and/or compositions containing them.
  • one or more polysaccharides are used to lower cholesterol, prevent sexually transmitted diseases, lubricate joints, regulate insulin levels, enhance cosmetics, stabilize or emulsify foods, and treat or effect prophylaxis of inflammation.
  • the invention includes a nutraceutical composition containing one or more polysaccharides disclosed herein and a carrier suitable for human consumption.
  • the composition contains the carrier and homogenized microalgae cells, such as red microalgae cells as a non-limiting example.
  • the composition contains the carrier and a purified first polysaccharide produced from a microalgal species listed in Table 1, which lists non-limiting examples of microalgae for the practice of the invention.
  • the carrier include a human nutritional supplement, such as vitamins, minerals, herbal extracts, monosaccharides or polysaccharides (e.g. glucosamine, glucosamine sulfate, chondroitin, or chondroitin sulfate, etc.) and proteins (e.g. protein supplements, etc.); a human food product; and various human foods perse.
  • the invention relates to compositions for topical application.
  • the composition is that of a cosmeceutical.
  • a cosmeceutical may contain one or more microalgal polysaccharides, or a microalgal cell homogenate, and a topical carrier.
  • the carrier may be any carrier suitable for topical application, such as, but not limited to, use on human skin or human mucosal tissue.
  • the composition may contain a purified microalgal polysaccharide, such as an exopolysaccharide, and a topical carrier.
  • the composition may contain a microalgal polysaccharide or homogenate and other component material found in cosmetics.
  • the component material may be that of a fragrance, a colorant (e.g. black or red iron oxide, titanium dioxide and/or zinc oxide, etc.), a sunblock (e.g. titanium, zinc, etc.), and a mineral or metallic additive.
  • the invention includes methods of preparing or producing a microalgal polysaccharide.
  • the invention includes methods that separate the exopolysaccharide from other molecules present in the medium used to culture exopolysaccharide producing microalgae.
  • separation includes removal of the microalgae from the culture medium containing the exopolysaccharide, after the microalgae has been cultured for a period of time.
  • the methods may be practiced with microalgal polysaccharides other than exopolysaccharides.
  • the methods include those where the microalgae was cultured in a bioreactor, optionally where a gas is infused into the bioreactor.
  • the invention includes a method of producing an exopolysaccharide, wherein the method comprises culturing microalgae in a bioreactor, wherein gas is infused into the bioreactor, separating the microalgae from culture media, wherein the culture media contains the exopolysaccharide; and separating the exopolysaccharide from other molecules present in the culture media.
  • the microalgae of the invention may be that of any species, including those listed in Table 1 herein.
  • the microalgae is a red algae, such as the red algae Porphyridium, which has two known species ⁇ Porphyridium sp. and Porphyridium cruentum) that have been observed to secrete large amounts of polysaccharide into their surrounding growth media.
  • the microalgae is of a genus selected from Rhodella, Chlorella, and ⁇ chnanthes.
  • Non-limiting examples of species within a microalgal genus of the invention include Porphyridium sp., Porphyridium cruentum, Porphyridium purpureum, Porphyridium aerugineum, Rhodella maculata, Rhodella reticulata, Chlorella autotrophica, Chlorella stigmatophora, Chlorella capsulata, Achnanthes brevipes and Achnanthes longipes.
  • a polysaccharide preparation method is practiced with culture media containing over 26.7, or over 27, rnM sulfate (or total SO 4 2" ).
  • Non-limiting examples include media with more than about 28, more than about 30, more than about 35, more than about 40, more than about 45, more than about 50, more than about 55, more than about 60, more than about 65, more than about 70, more than about 75, more than about 80, more than about 85, more than about 90, more than about 95, or more than about 100 mM sulfate.
  • more than about 150, more than about 200, more than about 250, more than about 300, more than about 350, more than about 400, more than about 450, more than about 500, more than about 550, more than about 600, more than about 650, more than about 700, or more than about 750, more than about 800, more than about 850, more than about 900, more than about 950, or more than about 1 M sulfate may be used to culture the microalgae and produce polysaccharides.
  • Sulfate in the media may be provided in one or more of the following forms: Na 2 SO 4 -IO H 2 O, MgSO 4 -7H 2 0, MnSO 4 , and CuSO 4 .
  • inventions of the method include the separation of an exopolysaccharide from other molecules present in the culture media by tangential flow filtration.
  • the methods may be practiced by separating an exopolysaccharide from other molecules present in the culture media by alcohol precipitation.
  • alcohols to use include ethanol, isopropanol, and methanol.
  • a method may further comprise treating a polysaccharide or exopolysaccharide with a protease to degrade polypeptide (or proteinaceous) material attached to, or found with, the polysaccharide or exopolysaccharide.
  • the methods may optionally comprise separating the polysaccharide or exopolysaccharide from proteins, peptides, and amino acids after protease treatment.
  • the invention includes methods of preparing a composition containing a microalgal polysaccharide or homogenate.
  • a method of producing a nutraceutical composition is described.
  • the composition may be prepared by drying a homogenate of microalgae after the microalgae have been disrupted to produce a homogenate.
  • the microalgae is separated from the culture medium used to grow the microalgae.
  • microalgae uses red microalgae to prepare the homogenate.
  • a homogenate processed as described herein may be combined with an appropriate carrier to form a nutraceutical of the invention.
  • the invention further includes a composition comprising a first component comprising cell material of the genus Porphyridium and at least one cholesterol lowering agent or compound.
  • the cell material is a cholesterol lowering homogenate, or a preparation of sulfated polysaccharides, and the agent or compound is selected from a plant phytosterol, a limonoid, a flavonoid, a tocotrienol, or any combination thereof.
  • the agent is an inhibitor of HMG CoA reductase, such as a statin.
  • the cell material may also be cells (biom&ss) per se, and in further embodiments, the cell material and cholesterol lowering agent may be packaged for sale as a single unit.
  • the method comprises culturing cells of the genus Porphyridium; separating the cells from culture media to prepare a cell containing material (biomass); drying the cell containing material; and mixing the dried material with at least one cholesterol lowering agent or compound.
  • the dried material is homogenized before mixing with the agent or compound.
  • the cells are homogenized before drying.
  • the agent or compound is selected from a plant phytosterol, a limonoid, a flavonoid, a tocotrienol, or a combination thereof, including a combination of two, three or all four thereof.
  • a method of formulating a cosmeceutical composition is disclosed.
  • the composition may be prepared by adding separated polysaccharides, or exopolysaccharides, to homogenized microalgal cells before, during, or after homogenization.
  • Both the polysaccharides and the microalgal cells may be from a culture of microalgae cells in suspension and under conditions allowing or permitting cell division.
  • the culture medium containing the polysaccharides is then separated from the microalgal cells followed by 1) separation of the polysaccharides from other molecules in the medium and 2) homogenization of the cells.
  • compositions of the invention may be formulated by subjecting a culture of microalgal cells and soluble exopolysaccharide to tangential flow filtration until the composition is substantially free of salts.
  • a polysaccharide is prepared after proteolysis of polypeptides present with the polysaccharide.
  • the polysaccharide and any contaminating polypeptides may be that of a culture medium separated from microalgal cells in a culture thereof. Ln some embodiments, the cells are of the genus Porphyridium.
  • the invention relates to methods of using a composition of the invention.
  • a method of lowering serum cholesterol is described.
  • the method may include orally administering, to a subject in need thereof, a polysaccharide produced by microalgae in combination with a biologically acceptable carrier.
  • a method is practiced by using a cholesterol lowering composition as described herein.
  • a composition contains a purified microalgal exopolysaccharide, or a microalgal cell homogenate, and a carrier suitable for human oral consumption.
  • a method of preventing a sexually transmitted disease includes administration of a solution comprising a polysaccharide produced by microalgae and use of a prophylactic device. In other embodiments, the solution is administered via the device.
  • a method of mammalian joint lubrication includes injecting polysaccharide produced by microalgae into a cavity containing synovial fluid.
  • a method of regulating insulin includes administering a polysaccharide produced by microalgae.
  • a method of cosmetic enhancement may include injecting a polysaccharide produced by microalgae into mammalian skin.
  • a method of stabilizing or emulsifying a food composition includes adding a polysaccharide produced by microalgae into a food composition.
  • a method of treating or effecting prophylaxis of mammalian inflammation includes administering a polysaccharide produced by microalgae to a mammal.
  • An additional aspect of the invention includes a method of reducing reactive oxygen species (ROS) in a mammal.
  • the method comprises administering a composition comprising disrupted red microalgae cells as described herein.
  • the composition comprises one or more other agents or compounds with antioxidant activity.
  • the method may be used to lower the level of ROS, or reduce or decrease the amount of damage caused by ROS.
  • a composition comprising homogenized red microalgae cells as described herein may also be used in additional methods of the invention.
  • the composition may be used in a method of delaying the onset of, or treating, a neurodegenerative disease in a mammal.
  • the neurodegenerative disease may be selected from Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, Freidreich's ataxia, Huntington's disease, or a Prion disease.
  • the composition may also be used in a method of reducing organ transplant rejection, a method of reducing the percentage of a mammalian body that is made of fat, a method of increasing satiety (or decreasing appetite) in a mammal, a method of increasing the energy expenditure of a mammal, a method of reducing the body weight of a mammal, a method of reducing inflammation in a mammal, or a method for the prevention and/or treatment of atherosclerosis.
  • Each of these methods may comprise administering a composition comprising homogenized red microalgae cells in a sufficient or effective amount as described herein.
  • the invention describes recombinant methods to modify microalgal cells.
  • the methods produce a microalgal cell that expresses an exogenous gene product.
  • the exogenous gene product may encode a carbohydrate transporter protein as a non-limiting example.
  • a microalgal cell containing an exogenous gene encoding a mammalian growth hormone is described.
  • the recombinantly modified cells per se, whether newly created or maintained in culture, are also part of the invention.
  • the invention also describes methods of recombinantly modifying a microalgal cell.
  • a method of trophically converting a microalgal cell such as members of the genus Porphyridium, is described.
  • the method may include selecting cells for a phenotype after transforming cells with a nucleic acid molecule in an expressible form, hi some methods, the phenotype may be the ability to undergo cell division in the absence of light and/or in the presence of a carbohydrate that is transported by a carbohydrate transporter protein encoded by the nucleic acid molecule.
  • the method may include use of an expression vector containing a nucleic acid sequence encoding a polypeptide, such as a carbohydrate transporter protein.
  • the method may include transforming a microalgal cell with a dual expression vector containing 1) a resistance cassette with a gene encoding a protein that confers resistance to an antibiotic, such as zeocin as a non-limiting example, operably linked to a promoter active in microalgae; and 2) a second expression cassette with a gene encoding a second protein operably linked to a promoter active in microalgae.
  • cells may be selected for the ability to survive in the presence of the antibiotic, such as at least 2.5 ⁇ g/ml zeocin as a non-limiting example where zeocin resistance is used.
  • the antibiotic can be at least 3.0 ⁇ g/ml zeocin, at least 4.0 ⁇ g/ml zeocin, at least 5.0 ⁇ g/ml zeocin, at least 6.0 ⁇ g/ml zeocin, at least 7.0 ⁇ g/ml zeocin, and at least 8.0 ⁇ g/ml zeocin.
  • the invention further relates to microalgal cells expressing a carbohydrate transporter protein for use in a method of producing a glycopolymer.
  • the method may include providing a transgenic cell containing an expressible gene encoding a monosaccharide transporter; and culruring the cell in the presence of at least one monosaccharide, transported into the cell by the transporter, wherein the monosaccharide is incorporated into a polysaccharide made by the cell.
  • a method of trophically converting a microalgae cell may include selecting for the ability to undergo cell division in the absence of light after subjecting the microalgal cell to a mutagen and placing the cell in the presence of a molecule listed in Tables 2 or 3 herein.
  • Another embodiment of the invention includes a recombinant nucleic acid comprising an endogenous promoter from Porphyridium sp. Also included in the invention is a recombinant promoter sequence that drives expression of the gene encoding the glycoprotein associated with the polysaccharide from Porphyridium sp., GenBank accession number AAV48590. In some embodiments the promoter or a segment thereof is operably linked to a coding region encoding a carbohydrate transporter or an antibiotic resistance gene.
  • the invention further comprises a cell of the genus Porphyridum, wherein the genome of the cell contains an exogenous gene encoding a carbohydrate transport protein.
  • the species is selected from the group consisting of Porphyridium sp. UTEX 637 or a strain derived from Porphyridium sp.
  • the carbohydrate transport protein has at least 60% amino acid identity with a member of the group consisting of SEQ ID NOs: 20, 22, 24, 26, 27, 29-39, and 46-48.
  • Figure 1 shows precipitation of 4 liters of Porphyridium cruentum exopolysaccharide using 38.5% isopropanol. (a) supernatant; (b) addition of 38.5% isopropanol; (c) precipitated polysaccharide; (d) separating step.
  • Figure 2 shows Porphyridium sp. cultured on agar plates containing various concentrations of zeocin.
  • Figure 3 shows growth of Porphyridium sp. and Porphyridium cruentum cells grown in light in the presence of various concentrations of glycerol.
  • Figure 4 shows Porphyridium sp. cells grown in the dark in the presence of various concentrations of glycerol.
  • Figure 5 shows levels of solvent-accessible polysaccharide in Porphyridium sp. homogenates subjected to various amounts of physical disruption from Sonication Experiment 1.
  • Figure 6 shows levels of solvent-accessible polysaccharide in Porphyridium sp. homogenates subjected to various amounts of physical disruption from Sonication Experiment 2.
  • Figure 7 shows sexually transmitted disease prevention devices containing various amounts of exopolysaccharide.
  • Figure 8 shows protein concentration measurements of autoclaved, protease-treated, and diafiltered exopolysaccharide.
  • Figure 9 shows various amounts and ranges of amounts of compounds found per gram of cells in cells of the genus Porphyridium.
  • Figure 10 (a) shows the diets used for animals of Groups 1 to 8.
  • Figure 10 (b) shows total serum cholesterol (TSC) levels (in mmol/L) in mice administered the diets of Groups 1 to 8.
  • Figure 10 (c) shows the mean TSC levels in mice administered the diets of Groups 1 to 8.
  • Figure 11 (a) shows the average food intake (grams) per day in mice administered the diets of Groups 1 to 8.
  • Figure 11 (b) shows the average body weight in mice administered the diets of Groups 1 to 8 over a 4 week period.
  • Figure 12 shows the percentage of total fat mass in mice administered the diets of Groups 1 to 8.
  • Figure 13 shows the energy expenditure in mice administered the diets of Groups 1 to 8.
  • Figure 14 shows the mean concentration of plasma antioxidants in mice administered the diets of Groups 1 to 8.
  • Figure 15(a) shows PCR genotyping of two Porphyridiurn transformants for the ble antibiotic resistance transgene.
  • Figure 15(b) shows PCR genotyping of two Porphyridium transformants for the endogenous glycoprotein gene promoter.
  • Figure 15(c) shows PCR genotyping of a selected Porphyridium transformant for an exogenous gene encoding a recoded human GLUTl transporter.
  • Figure 16 shows a Southern blot indicating chromosomal integration of an exogenous gene encoding a recoded human GLUTl transporter.
  • This application is a continuation-in-part of and claims priority to U.S. Patent application No:l 1/336,428, filed January 19, 2006, entitled “Methods and Compositions for Cholesterol Reduction in Mammals", which is hereby incorporated in its entirety for all purposes.
  • This application is a continuation-in-part of and claims priority to U.S. Patent application No: 11/337,171, filed January 19, 2006, entitled “Methods and Compositions for Reducing Inflammation and Preventing Oxidative Damage", which is hereby incorporated in its entirety for all purposes.
  • This application is a nonprovisional of and claims priority to U.S. Patent application No: 60/838,452, filed August 17, 2006, entitled “Polysaccharide Compositions and Methods of Administering, Producing, and Formulating Polysaccharide Compositions", which is hereby incorporated in its entirety for all purposes.
  • This application is a nonprovisional of and claims priority to U.S.
  • This application is a nonprovisional of and claims priority to U.S. Patent application No: 60/872,072, filed November 30, 2006, entitled “Polysaccharide Compositions and Methods of Administering, Producing, and Formulating Polysaccharide Compositions", which is hereby incorporated in its entirety for all purposes.
  • U.S. Patent application No: filed January 19,
  • “Active in microalgae” means a nucleic acid that is functional in microalgae.
  • a promoter that has been used to drive an antibiotic resistance gene to impart antibiotic resistance to a transgenic microalgae is active in microalgae.
  • Nonlimiting examples of promoters active in microalgae are promoters endogenous to certain algae species and promoters found in plant viruses.
  • Antiviral lubricant means a molecule that possesses both antiviral activity and lubricant activity.
  • ARA Arachidonic acid
  • “Associates with” means, within the context of a polysaccharide binding fusion protein, one molecule binding to another molecule. Affinity and selectivity of binding can vary when a polysaccharide and a polysaccharide binding protein are in association with each other. "Axenic” means a culture of an organism that is free frqm contamination by other living organisms.
  • Bioreactor means an enclosure or partial enclosure in which cells are cultured in suspension.
  • Carbohydrate modifying enzyme means an enzyme that utilizes a carbohydrate as a substrate and structurally modifies the carbohydrate.
  • Carbohydrate transporter means a polypeptide that resides in a lipid bilayer and facilitates the transport of carbohydrates across the lipid bilayer.
  • Carrier suitable for human consumption refers to compounds and materials suitable for human ingestion or otherwise physiologically compatible with oral administration to humans. Usually, such carriers are of plant or animal origin. Although such carriers sometimes contain residual amounts of solvents and buffers used in the processing of the polysaccharides and other compositions of the invention, they do not consist exclusively of such solvents or buffers, and usually have less than 50% and preferably less than 10% w/w of such solvents or buffers.
  • Carrier suitable for topical administration means a compound that may be administered, together with one or more compounds of the present invention, and which does not destroy the activity thereof and is nontoxic when administered in concentrations and amounts sufficient to deliver the compound to the skin or a mucosal tissue.
  • Cell material means any matter obtained from cells.
  • Cell material can be intact cells, disrupted cells, a purified componenet of cells, and extract of cells, and other materials.
  • Combination Product means a product that comprises at least two distinct compositions intended for human administration through distinct routes, such as a topical route and an oral route. In some embodiments the same active agent is contained in both the topical and oral components of the combination product.
  • “Conditions favorable to cell division” means conditions in which cells divide at least once every 72 hours.
  • “Derived from” means, within the context of a microalgae strain, a strain that has been generated through passaging or mutagenesis from a starting strain.
  • Porphyridium strain UTEX 637 can be subjected to chemical mutagenesis to generate new colonies. Each new colony is a strain deruived from Porphyridium strain UTEX 637.
  • DHA Docosahexaenoic acid
  • Endopolysaccharide means a polysaccharide that is retained intracellularly.
  • EPA means eicosapentaenoic acid.
  • Exogenous gene means agene transformed into a wild-type organism.
  • the gene can be heterologous from a different species, or homologous from the same species, in which case the gene occupies a different location in the genome of the organism than the endogenous gene.
  • Exogenously provided describes a molecule provided to the culture media of a cell culture.
  • Exopolysaccharide means a polysaccharide that is secreted from a cell into the extracellular environment.
  • Frtrate means the portion of a tangential flow filtration sample that has passed through the filter.
  • Fiberd carbon source means molecule(s) containing carbon that are present at ambient temperature and pressure in solid or liquid form.
  • glycopolymer means a biologically produced molecule comprising at least two monosaccharides.
  • examples of glycopolymers include glycosylated proteins, polysaccharides, oligosaccharides, and disaccharides.
  • a coding sequence is described as "heterologous" when it is physically connected to a nucleic acid other than that which it is naturally connected to in a wild-type genome.
  • a coding sequence not found in a wild-type genome is necessarily heterologous to a promoter that it is in operable linkage with.
  • Homogenate means cell biomass that has been disrupted.
  • a homogenate is not necessarily completely homogeneous, and as used herein can refer to a composition of cells that have been disrupted to the point where a majority of the cells in the preparation have been ruptured.
  • a homogenate may contain chunks of cell wall, intact or partially intact organelles such as chloroplasts and mitochondria, and other substructures of the cell.
  • Microalgae means a single-celled organism that is capable of performing photosynthesis. Microalgae include obligate photoautotrophs, which cannot metabolize a fixed carbon source as energy, as well as heterotrophs, which can live solely off of light, solely off of a fixed carbon source, or a combination of the two.
  • “Naturally produced” describes a compound that is produced by a wild-type organism.
  • “Pharmaceutically acceptable carrier or adjuvant” refers to a carrier or adjuvant that may be administered to a patient, together with one or more compounds of the present invention, and which does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the compound.
  • “Photobioreactor” means a waterproof container, at least part of which is at least partially transparent, allowing light to pass through, in which one or more microalgae cells are cultured. Photobioreactors may be sealed, as in the instance of a polyethylene bag, or may be open to the environment, as in the instance of a pond.
  • Polysaccharide material is a composition that contains more than one species of polysaccharide, and optionally contaminants such as proteins, lipids, and nucleic acids, such as, for example, a microalgal cell homogenate.
  • Polysaccharide means a compound or preparation containing one or more molecules that contain at least two saccharide molecules covalently linked.
  • a “polysaccharide”, “endopolysaccharide” or “exopolysaccharide” can be a preparation of polymer molecules that have similar or identical repeating units but different molecular weights within the population.
  • Port in the context of a photobioreactor, means an opening in the photobioreactor that allows influx or efflux of materials such as gases, liquids, and cells. Ports are usually connected to tubing leading to and/or from the photobioreactor.
  • Red microalgae means unicellular algae that is of the list of classes comprising Bangiophyceae, Florideophyceae, Goniotrichales, or is otherwise a member of the Rhodophyta.
  • Retentate means the portion of a tangential flow filtration sample that has not passed through the filter.
  • “Selectively binds to” refers to a binding reaction which is determinative of the presence of a molecule in the presence of a heterogeneous population of other molecules. Thus, under designated conditions, a specified ligand binds preferentially to a particular molecule and does not bind in a significant amount to other proteins present in the sample.
  • a molecule such as antibody that specifically binds to a protein often has an association constant of at least 10 6 M '1 or 10 7 M "1 , preferably 10 8 M “1 to 10 9 M '1 , and more preferably, about 10 10 M "1 to 10 1 ' M "1 or higher.
  • a variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein.
  • solid- phase ELISA immunoassays are routinely used to select monoclonal antibodies specifically immunoreactive with a protein. See, e.g., Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, New York, for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity.
  • Small molecule means a molecule having a molecular weight of less than 2000 daltons, in some instances less than 1000 daltons, and in still other instances less than 500 daltons or less. Such molecules include, for example, heterocyclic compounds, carbocyclic compounds, sterols, amino acids, lipids, carotenoids and polyunsaturated fatty acids.
  • a molecule is "solvent available” when the molecule is isolated to the point at which it can be dissolved in a solvent, or sufficiently dispersed in suspension in the solvent such that it can be detected in the solution or suspension.
  • a polysaccharide is “solvent available” when it is sufficiently isolated from other materials, such as those with which it is naturally associated, such that the polysaccharide can be dissolved or suspended in an aqueous buffer and detected in solution using a dimethylmethylene blue (DMMB) or phenohsulfuric acid assay.
  • DMMB dimethylmethylene blue
  • part of the polysaccharide can be "solvent available” when it is on the outermost layer of a cell wall while other parts of the same polysaccharide molecule are not “solvent available” because they are buried within the cell wall.
  • solvent available polysaccharide since most of the cell wall polysaccharide is sequestered within the cell wall and not available to solvent.
  • the amount of "solvent available” polysaccharide increases.
  • the amount of "solvent accessible" polysaccharide before and after homogenization can be compared by taking two aliquots of equal volume of cells from the same culture, homogenizing one aliquot, and comparing the level of polysaccharide in solvent from the two aliquots using a DMMB assay.
  • the amount of solvent accessible polysaccharide in a homogenate of cells can also be compared with that present in a quantity of cells of the same type in a different culture needed to generate the same amount of homogenate.
  • “Substantially free of protein” means compositions that are preferably of high purity and are substantially free of potentially harmful contaminants, including proteins (e.g., at least National Food (NF) grade, generally at least analytical grade, and more typically at least pharmaceutical grade).
  • compositions are at least 80, at least 90, at least 99 or at least 99.9% w/w pure of undesired contaminants such as proteins are substantially free of protein. To the extent that a given compound must be synthesized prior to use, the resulting product is typically substantially free of any potentially toxic agents, particularly any endotoxins, which may be present during the synthesis or purification process.
  • Compositions are usually made under GMP conditions.
  • Compositions for parenteral administration are usually sterile and substantially isotonic.
  • Polysaccharides form a heterogeneous group of polymers of different length and composition. They are constructed from monosaccharide residues that are linked by glycosidic bonds. Glycosidic linkages may be located between the Ci (or C 2 ) of one sugar residue and the C 2 , C 3 , C 4 , C 5 or Ce of the second residue. A branched sugar results if more than two types of linkage are present in single monosaccharide molecule.
  • Monosaccharides are simple sugars with multiple hydroxyl groups. Based on the number of carbons (e.g., 3, 4, 5, or 6) a monosaccharide is a triose, tetrose, pentose, or hexose. Pentoses and hexoses can cyclize, as the aldehyde or keto group reacts with a hydroxyl on one of the distal carbons. Examples of monosaccharides are galactose, glucose, and rhamnose.
  • Polysaccharides are molecules comprising a plurality of monosaccharides covalently linked to each other through glycosidic bonds. Polysaccharides consisting of a relatively small number of monosaccharide units, such as 10 or less, are sometimes referred to as oligosaccharides. The end of the polysaccharide with an anomeric carbon (Ci) that is not involved in a glycosidic bond is called the reducing end.
  • a polysaccharide may consist of one monosaccharide type, known as a homopolymer, or two or more types of monosaccharides, known as a heteropolymer.
  • homopolysaccharides are cellulose, amylose, inulin, chitin, chitosan, amylopectin, glycogen, and pectin.
  • Amylose is a glucose polymer with ⁇ (l ->4) glycosidic linkages.
  • Amylopectin is a glucose polymer with ⁇ (l ->4) linkages and branches formed by ⁇ (l ->6) linkages.
  • heteropolysaccharides are glucomannan, galactoglucomannan, xyloglucan, 4-O- methylglucuronoxylan, arabinoxylan, and 4-O-Methylglucuronoarabinoxylan.
  • Polysaccharides can be structurally modified both enzymatically and chemically. Examples of modifications include sulfation, phosphorylation, methylation, O-acetylation, fatty acylation, amino N-acetylation, N-sulfation, branching, and carboxyl lactonization.
  • Glycosaminogl yeans are polysaccharides of repeating disaccharides. Within the disaccharides, the sugars tend to be modified, with acidic groups, amino groups, sulfated hydroxyl and amino groups. Glycosaminoglycans tend to be negatively charged, because of the prevalence of acidic groups. Examples of glycosaminoglycans are heparin, chondroitin, and hyaluronic acid.
  • Polysaccharides are produced in eukaryotes mainly in the endoplasmic reticulum (ER) and Golgi apparatus. Polysaccharide biosynthesis enzymes are usually retained in the ER, and amino acid motifs imparting ER retention have been identified (Gene. 2000 Dec 31;261(2):321-7). Polysaccharides are also produced by some prokaryotes, such as lactic acid bacteria.
  • Polysaccharides that are secreted from cells are known as exopolysaccharides.
  • Many types of cell walls, in plants, algae, and bacteria are composed of polysaccharides. The cell walls are formed through secretion of polysaccharides.
  • Some species, including algae and bacteria secrete polysaccharides that are released from the cells. In other words, these molecules are not held in association with the cells as are cell wall polysaccharides. Instead, these molecules are released from the cells. For example, cultures of some species of microalgae secrete exopolysaccharides that are suspended in the culture media.
  • Polysaccharides can be produced by culturing microalgae. Examples of microalgae that can be cultured to produce polysaccharides are shown in Table 1. Also listed are references that enable the skilled artisan to culture the microalgae species under conditions sufficient for polysaccharide production. Also listed are strain numbers from various publicly available algae collections, as well as strains published in journals that require public dissemination of reagents as a prerequisite for publication.
  • Microalgae are preferably cultured in liquid media for polysaccharide production. Culture condition parameters can be manipulated to optimize total polysaccharide production as well as to alter the structure of polysaccharides produced by microalgae.
  • Microalgal culture media usually contains components such as a fixed nitrogen source, trace elements, a buffer for pH maintenance, and phosphate.
  • Other components can include a fixed carbon source such as acetate or glucose, and salts such as sodium chloride, particularly for seawater microalgae.
  • trace elements include zinc, boron, cobalt, copper, manganese, and molybdenum in, for example, the respective forms of ZnCl 2 , H 3 BO 3 , CoCl 2 -6H 2 O, CuCl 2 -2H 2 O, MnCl 2 -4H 2 O and (NH 4 ) 6 Mo 7 ⁇ 24 -4H 2 O.
  • microalgae species can grow by utilizing a fixed carbon source such as glucose or acetate. Such microalgae can be cultured in bioreactors that do not allow light to enter. Alternatively, such microalgae can also be cultured in photobioreactors that contain the fixed carbon source and allow light to strike the cells. Such growth is known as heterotrophic growth. Any strain of microalgae, including those listed in Table 1, can be cultured in the presence of any one or more fixed carbon source including those listed in Tables 2 and 3.
  • a fixed carbon source such as glucose or acetate
  • 6-deoxy-6-fl ⁇ xorosucrose ⁇ -deoxy-alpha-gluco-pyranosiduronic acid ⁇ -deoxy-gluco-heptopyranosyl ⁇ -deoxy-gluco-heptopyranoside
  • Microalgae contain photosynthetic machinery capable of metabolizing photons, and transferring energy harvested from photons into fixed chemical energy sources such as monosaccharide.
  • Glucose is a common monosaccharide produced by microalgae by metabolizing light energy and fixing carbon from carbon dioxide. Some microalgae can also grow in the absence of light on a fixed carbon source that is exogenously provided (for example see Plant Physiol. 2005 Feb;137(2):460-74).
  • monosaccharides such as glucose are also substrate for production of polysaccharides (see Example 14).
  • the invention provides methods of producing polysaccharides with novel monosaccharide compositions.
  • microalgae is cultured in the presence of culture media that contains exogenously provided monosaccharide, such as glucose.
  • the monosaccharide is taken up by the cell by either active or passive transport and incorporated into polysaccharide molecules produced by the cell.
  • This novel method of polysaccharide composition manipulation can be performed with, for example, any microalgae listed in Table 1 using any monosaccharide or disaccharide listed in Tables 2 or 3.
  • the fixed carbon source is one or more selected from glucose, galactose, xylose, mannose, rhamnose, N-acetylglucosamine, glycerol, floridoside, and glucuronic acid.
  • the methods may be practiced cell growth in the presence of at least about 5.0 ⁇ M, at least about 10 ⁇ M, at least about 15.0 ⁇ M, at least about 20.0 ⁇ M, at least about 25.0 ⁇ M, at least about 30.0 ⁇ M, at least about 35.0 ⁇ M, at least about 40.0 ⁇ M, at least about 45.0 ⁇ M, at least about 50.0 ⁇ M, at least about 55.0 ⁇ M, at least about 60.0 ⁇ M, at least about 75.0 ⁇ M, at least about 80.0 ⁇ M, at least about 85.0 ⁇ M, at least about 90.0 ⁇ M, at least about 95.0 ⁇ M, at least about 100.0 ⁇ M, or at least about 150.0 ⁇ M, of one or more exogenously provided fixed carbon sources selected from Tables 2 and 3.
  • the methods include the use of approximately 0.5-0.75% glycerol as a fixed carbon source when the cells are cultured in the presence of light.
  • a range of glycerol from approximately 4.0% to approximately 9.0% may be used when the Porphyridium cells are cultured in the dark, more preferably from 5.0% to 8.0%, and more preferably 7.0%.
  • the monosaccharide composition of the polysaccharide can be analyzed as described in Example 5.
  • Microalgae can be transformed with genes encoding carbohydrate transporters to facilitate the uptake of exogenously provided carbohydrates such SEQ ID NOs: 20, 22, 24, 26, 27, 29-39 and 46-48.
  • Microalgae culture media can contain a fixed nitrogen source such as KNO 3 .
  • microalgae are placed in culture conditions that do not include a fixed nitrogen source.
  • Porphyridium sp. cells are cultured for a first period of time in the presence of a fixed nitrogen source, and then the cells are cultured in the absence of a fixed nitrogen source (see for example Adda M., Biomass 10:131-140. (1986); Sudo H., et al., Current Microbiology Vol. 30 (1995), pp. 219-222; Marinho-Soriano E., Bioresour Technol. 2005 Feb;96(3):379-82; Bioresour. Technol. 42:141-147 (1992)).
  • culture parameters can also be manipulated, such as the pH of the culture media, the identity and concentration of trace elements such as those listed in Table 3, and other media constituents.
  • One non-limiting example is the inclusion of at least one source of sulfate in the culture media to increase the level of sulfation in the polysaccharides produced.
  • a polysaccharide preparation method is practiced with culture media containing more than about 100, more than about 150, more than about 200, more than about 250, more than about 300, more than about 350, more than about 400, more than about 450, more than about 500, more than about 550, more than about 600, more than about 650, more than about 700, or more than about 750, more than about 800, more than about 850, more than about 900, more than about 950, or more than about 1 or 2 M sulfate (or total SO 4 2" ).
  • Increasing the sulfation has been demonstrated to increase the antioxidant apacity of the polysaccharide (see example 23 and Spitz et al. (J. Appl.
  • the invention is based in part on the surprising discovery that growth under higher sulfate concentrations can produce polysaccharides with higher levels of sulfation. This allows for the production of cells (and so biomass) containing highly sulfated polysaccharides that may be used in the form of purified polysaccharides, a homogenate of cells (biomass), intact cells (biomass) per se, or a combination thereof.
  • Porphyridiiim can survive above 10OmM sulfate was surprising for a number of reasons.
  • sulfate can alter the rate of uptake of toxic metal ions in algae, such as chromium, and that increasing metal accumulation can lead to toxicity (see for example Kaszycki et al, Plant, Cell & Environment, 28(2): p.260, 2005).
  • sulfate can inhibit the uptake of metal ions required for nitrogen fixation such as molybdenum, and that increasing sulfate concentrations negativelky affects algae such as cyanobacteria even at sulfate concentrations in estuarine (>8— 10 mM) and seawater (28mM) levels of sufate (see for example Marino et al, Hydrobiologia 500: pp. 277-293, 2004).
  • sulfate at high levels can often be taken up and reduced and sulfide, which is is toxic to photosynthesis because it attacks photosystem II (see for example Khanal et al., J. Envir. Engrg., 129(12); pp.
  • Microalgae can be grown in the presence of light. The number of photons striking a culture of microalgae cells can be manipulated, as well as other parameters such as the wavelength spectrum and ratio of darkdight hours per day. Microalgae can also be cultured in natural light, as well as simultaneous and/or alternating combinations of natural light and artificial light. For example, microalgae of the genus Chlorella be cultured under natural light during daylight hours and under artificial light during night hours.
  • the gas content of a photobioreactor can be manipulated. Part of the volume of a photobioreactor can contain gas rather than liquid. Gas inlets can be used to pump gases into the photobioreactor. Any gas can be pumped into a photobioreactor, including air, air/C ⁇ 2 mixtures, noble gases such as argon and others. The rate of entry of gas into a photobioreactor can also be manipulated. Increasing gas flow into a photobioreactor increases the turbidity of a culture of microalgae. Placement of ports conveying gases into a photobioreactor can also affect the turbidity of a culture at a given gas flow rate. Air/CO 2 mixtures can be modulated to generate different polysaccharide compositions by manipulating carbon flux.
  • air:CO 2 mixtures of about 99.75% air:0.25% CO2, about 99.5% air:0.5% CO 2 , about 99.0% air: 1.00% CO 2 , about 98.0% air:2.0% CO 2 , about 97.0% air:3.0% CO 2 , about 96.0% air:4.0% CO 2 , and about 95.00% air:5.0% CO 2 can be infused into a bioreactor or photobioreactor.
  • Microalgae cultures can also be subjected to mixing using devices such as spinning blades and propellers, rocking of a culture, stir bars, and other instruments.
  • Microalgae can be grown and maintained in closed photobioreactors made of different types of transparent or semitransparent material. Such material can include Plexiglas ® enclosures, glass enclosures, bags bade from substances such as polyethylene, transparent or semitransparent pipes, and other materials. Microalgae can also be grown in open ponds.
  • Photobioreactors can have ports allowing entry of gases, solids, semisolids and liquids into the chamber containing the microalgae. Ports are usually attached to tubing or other means of conveying substances. Gas ports, for example, convey gases into the culture. Pumping gases into a photobioreactor can serve to both feed cells CO 2 and other gases and to aerate the culture and therefore generate turbidity. The amount of turbidity of a culture varies as the number and position of gas ports is altered. For example, gas ports can be placed along the bottom of a cylindrical polyethylene bag. Microalgae grow faster when CO 2 is added to air and bubbled into a photobioreactor.
  • a 5% CO2'.95% air mixture is infused into a photobioreactor containing cells of the genus Porphyridium (see for example Biotechnol Bioeng. 1998 Sep 20;59(6):705-13; textbook from office; US Patents 5,643,585 and 5,534,417; Lebeau, T., et. al. Appl. Microbiol Biotechnol (2003) 60:612-623; Muller-Fuega, A., Journal of Biotechnology 103 (2003 153-163; Muller-Fuega, A., Biotechnology and Bioengineering, Vol. 84, No. 5, December 5, 2003; Garcia- Sanchez, J.L., Biotechnology and Bioengineering, Vol. 84, No. 5, December 5, 2003; Garcia- Gonzales, M., Journal of Biotechnology, 115 (2005) 81-90. Molina Grima, E., Biotechnology Advances 20 (2003) 491-515).
  • Photobioreactors can be exposed to one or more light sources to provide microalgae with light as an energy source via light directed to a surface of the photobioreactor.
  • the light source provides an intensity that is sufficient for the cells to grow, but not so intense as to cause oxidative damage or cause a photoinhibitive response.
  • a light source has a wavelength range that mimics or approximately mimics the range of the sun. In other instances a different wavelength range is used.
  • Photobioreactors can be placed outdoors or in a greenhouse or other facility that allows sunlight to strike the surface.
  • Preferred photon intensities for species of the genus Porphyridium are between 50 and 300 uE m '2 s '1 (see for example Photosynth Res. 2005 Jun;84(l-3):21-7).
  • Photobioreactor preferably have one or more parts that allow media entry. It is not necessary that only one substance enter or leave a port.
  • a port can be used to flow culture media into the photobioreactor and then later can be used for sampling, gas entry, gas exit, or other purposes.
  • a photobioreactor is filled with culture media at the beginning of a culture and no more growth media is infused after the culture is inoculated.
  • the microalgal biomass is cultured in an aqueous medium for a period of time during which the microalgae reproduce and increase in number; however quantities of aqueous culture medium are not flowed through the photobioreactor throughout the time period.
  • aqueous culture medium is not flowed through the photobioreactor after inoculation.
  • culture media can be flowed though the photobioreactor throughout the time period during which the microalgae reproduce and increase in number.
  • media is infused into the photobioreactor after inoculation but before the cells reach a desired density.
  • a turbulent flow regime of gas entry and media entry is not maintained for reproduction of microalgae until a desired increase in number of said microalgae has been achieved, but instead a parameter such as gas entry or media entry is altered before the cells reach a desired density.
  • Photobioreactors preferably have one or more ports that allow gas entry. Gas can serve to both provide nutrients such as CO 2 as well as to provide turbulence in the culture media. Turbulence can be achieved by placing a gas entry port below the level of the aqueous culture media so that gas entering the photobioreactor bubbles to the surface of the culture. One or more gas exit ports allow gas to escape, thereby preventing pressure buildup in the photobioreactor. Preferably a gas exit port leads to a "one-way" valve that prevents contaminating microorganisms to enter the photobioreactor.
  • cells are cultured in a photobioreactor for a period of time during which the microalgae reproduce and increase in number, however a turbulent flow regime with turbulent eddies predominantly throughout the culture media caused by gas entry is not maintained for all of the period of time. In other instances a turbulent flow regime with turbulent eddies predominantly throughout the culture media caused by gas entry can be maintained for all of the period of time during which the microalgae reproduce and increase in number. In some instances a predetermined range of ratios between the scale of the photobioreactor and the scale of eddies is not maintained for the period of time during which the microalgae reproduce and increase in number. In other instances such a range can be maintained.
  • Photobioreactors preferably have at least one port that can be used for sampling the culture.
  • a sampling port can be used repeatedly without altering compromising the axenic nature of the culture.
  • a sampling port can be configured with a valve or other device that allows the flow of sample to be stopped and started.
  • a sampling port can allow continuous sampling.
  • Photobioreactors preferably have at least one port that allows inoculation of a culture. Such a port can also be used for other purposes such as media or gas entry.
  • Microalgae that produce polysaccharides can be cultured in photobioreactors.
  • Microalgae that produce polysaccharide that is not attached to cells can be cultured for a period of time and then separated from the culture media and secreted polysaccharide by methods such as centrifugation and tangential flow filtration.
  • Preferred organisms for culturing in photobioreactors to produce polysaccharides include Porphyridium sp., Porphyridium cruentum, Porphyridium purpureum, Porphyridium aerugineum, Rhodella maculata, Rhodella reticulata, Chlorella autotrophica, Chlorella stigmatophora, Chlorella capsulata, Achnanthes brevipes and Achnanthes longipes.
  • Organisms besides microalgae can be used to produce polysaccharides, such as lactic acid bacteria (see for example Stinglee, F., Molecular Microbiology (1999) 32(6), 1287-1295; Ruasjvladiedo, P., J. Dairy Sci. 88:843-856 (2005); Laws, A-, Biotechnology Advances 19 (2001) 597-625; Xanthan gum bacteria: Pollock, TJ., J. Ind. Microbiol Biotechnol., 1997 Aug; 19(2): 92-7.; Becker, A., Appl. Micrbiol. Bioltechnol.
  • Microalgae and other organisms can be manipulated to produce polysaccharide molecules that are not naturally produced by methods such as feeding cells with monosaccharides that are not produced by the cells (Nature. 2004 Aug 19;430(7002):873- 7).
  • species listed in Table 1 are grown according to the referenced growth protocols, with the additional step of adding to the culture media a fixed carbon source that is not in the culture media as published and referenced in Table 1 and is not produced by the cells in a detectable amount.
  • such cells can first be transformed to contain a carbohydrate transporter, thus facilitating the entry of monosaccharides.
  • Polysaccharides can be altered by enzymatic and chemical modification.
  • carbohydrate modifying enzymes can be added to a preparation of polysaccharide and allowed to catalyze reactions that alter the structure of the polysaccharide.
  • Chemical methods can be used to, for example, modify the sulfation pattern of a polysaccharide (see for example Carbohydr. Polym. 63:75-80 (2000); Pomin VH., Glycobiology. 2005 Dec;15(12):1376-85; Naggi A., Semin Thromb Hemost. 2001 Oct;27(5):437-43 Review; Habuchi, O., Glycobiology. 1996 Jan;6(l);51-7; Chen, J., J. Biol. Chem. In press; Geresh., S et al., J. Biochem. Biophys. Methods 50 (2002) 179-187.).
  • Exopolysaccharides can be purified from microalgal cultures by various methods, including those disclosed herein.
  • polysaccharides can be precipitated by adding compounds such as cetylpyridinium chloride, isopropanol, ethanol, or methanol to an aqueous solution containing a polysaccharide in solution.
  • Pellets of precipitated polysaccharide can be washed and resuspended in water, buffers such as phosphate buffered saline or Tris, or other aqueous solutions (see for example Farias, W.R.L., et al., J. Biol. Chem. (2000) 275;(38)29299-29307; U.S. Patent No. 6,342,367; U.S. Patent No. 6,969,705).
  • Dialysis Polysaccharides can also be dialyzed to remove excess salt and other small molecules (see for example Gloaguen, V., et al., Carbohydr Res. 2004 Jan 2;339(l):97-103; Microbiol Immunol. 2000;44(5):395-400.).
  • Tangential Flow Filtration can be used to concentrate polysaccharide and remove salts.
  • tangential flow filtration also known as cross-flow filtration, can be used
  • For a preferred filtration method see Geresh, Carb. Polym. 50; 183-189 (2002), which discusses use of a MaxCell A/G technologies 0.45uM hollow fiber filter.
  • Millipore Pellicon ® devices used with 10OkD, 30OkD, 1000 kD (catalog number P2C01MC01), 0.IuM (catalog number P2VVPPV01), 0.22uM (catalog number P2GVPPV01), and 0.45uM membranes (catalog number P2HVMPV01). It is preferred that the polysaccharides do not pass through the filter at a significant level. It is also preferred that polysaccharides do not adhere to the filter material. TFF can also be performed using hollow fiber filtration systems.
  • Non-limiting examples of tangential flow filtration include use of a filter with a pore size of at least about 0.1 micrometer, at least about 0.12 micrometer, at least about 0.14 micrometer, at least about 0.16 micrometer, at least about 0.18 micrometer, at least about 0.2 micrometer, at least about 0.22 micrometer, or at least about 0.45 micrometer.
  • Preferred pore sizes of TFF allow contaminants to pass through but not polysaccharide molecules.
  • Tangential flow filtration may also be used to isolate and/or purify polysaccharides for further analysis or comparison.
  • the level of sulfation in polysaccharides from different cells or cell material may be analyzed or compared.
  • the cells or material may be resuspended in water or other suitable liquid to allow the release of polysaccharides into the liquid.
  • the release may be augmented by agitation of the cells or material in the liquid, such as by vortexing, or by the use of an elevated temperature, such as a warm liquid.
  • the released polysaccharides may be separated from the cells or insoluble cellular material by any convenient means, like centrifugation or filtration as non-limiting examples.
  • the polysaccharide containing liquid may then be subjected to tangential flow filtration to isolate polysaccharides suitable for further analysis, such as combustion to determine the amount of sulfation in the polysaccharides.
  • the amount of polysaccharide, or sulfation thereof may be standardized against any known cellular metabolite or component, such as, but not limited to, the amount of DNA per gram of cells or cell material.
  • the polysaccharides from a cell extract may be analyzed as described above. Polysaccharides released into the extract, optionally with agitation and/or an elevated temperature as disclosed herein, may be separated from insoluble cellular material as described above. The soluble material may then be subjected to tangential flow filtration to isolate polysaccharides for further analysis or comparison.
  • the amount or level of sulfation in the polysaccharides is analyzed and compared to the amount of sulfates used to culture the microalgae.
  • the amount or level of sulfation in the polysaccharides of cells grown at about 100 mM, about 200 mM, about 300 mM, about 400 mM, about 500 mM, about 600 mM, about 700 mM, or about 800 mM or higher, sulfate may be determined by routine and repetitive methods.
  • a disclosed polysaccharide may contain at least about 4.6% sulfur, at least about 4.75% sulfur, at least about 5.0% sulfur, at least about 5.25% sulfur, at least about 5.5% sulfur, at least about 5.75% sulfur, at least about 6.0% sulfur, at least about 6.25% sulfur, at least about 6.5% sulfur, at least about 6.75% sulfur, or at least about 7.0% sulfur by weight.
  • the amount or level of sulfur by weight in the polysaccharides of a sample of cells or cell material may determined without knowledge of the amount of sulfate used to culture the cells.
  • the amount or level of sulfur by weight may range from about 3.0 to about 7.0% as described above.
  • the amount or level of sulfur may be used in a method to determine the amount of sulfates used to culture the cells and produce the cell material. The method may comprise comparison of the amount or level of sulfur, per unit of cellular metabolite or component, in a sample to that of cells grown in known amounts of sulfates.
  • the method may be used to identify cells, and so polysaccharides, prepared or obtained by methods disclosed herein.
  • homogenized Porphyrdium biomass may be incubated in distilled water, subjected to agitation to realease polysaccharide into solution, and cenytrifiged to remove solids.
  • the resulting soluble polysaccharide can then be purified, concentrated, and diafiltered using methods disclosed in the Examples.
  • the resulting ghigh purity polysaccharide may then be subjected to sulfur content analysis.
  • the sulfur content alnalysis not only indicated the level of sulfation of the polysaccharide but also can indicate the level of sulfate under which the cells were cultured.
  • polysaccharides can be treated with proteases to degrade contaminating proteins.
  • the contaminating proteins are attached, either covalently or noncovalently to polysaccharides.
  • the polysaccharide molecules are in a preparation that also contains proteins.
  • Proteases can be added to polysaccharide preparations containing proteins to degrade proteins (for example, the protease from Streptomyces griseus can be used (SigmaAldrich catalog number P5147).
  • the polysaccharide is preferably purified from residual proteins, peptide fragments, and amino acids. This purification can be accomplished, for example, by methods listed above such as dialysis, filtration, and precipitation.
  • Heat treatment can also be used to eliminate proteins in polysaccharide preparations (see for example Biotechnol Lett. 2005 Jan;27(l):13-8; FEMS Immunol Med Microbiol. 2004 Oct 1;42(2): 155-66; Carbohydr Res. 2000 Sep 8;328(2):199-207; J Biomed Mater Res. 1999;48(2):l l l-6.; Carbohydr Res. 1990 Oct 15;207(l):101-20; ).
  • the invention thus includes production of an exopolysaccharide comprising separating the exopolysaccharide from contaminants after proteins attached to the exopolysaccharide have been degraded or destroyed.
  • the proteins may be those attached to the exopolysaccharide during culture of a microalgal cell in media, which is first separated from the cells prior to proteolysis or protease treatment.
  • the cells may be those of the genus Porphyridium as a non-limiting example.
  • a method of producing an exopolysaccharide comprising culturing cells of the genus Porphyridium; separating cells from culture media; destroying protein attached to the exopolysaccharide present in the culture media; and separating the exopolysaccharide from contaminants.
  • the contaminant(s) are selected from amino acids, peptides, proteases, protein fragments, and salts.
  • the contaminant is selected from NaCl, MgSO 4 , MgCl 2 , CaCl 2 , KNO 3 , KH 2 PO 4 , NaHCO 3 , Tris, ZnCl 2 , H 3 BO 3 , CoCl 2 , CuCl 2 , MnCl 2 , (NH 4 ) 6Mo 7 O 24 , FeC13 and EDTA.
  • polysaccharides can be dried using methods such as lyophilization and heat drying (see for example Shastry, S., Brazilian Journal of Microbiology (2005) 36:57-62; Matthews KH.Jnt J Pharm. 2005 Jan 31;289(l-2):51-62. Epub 2004 Dec 30; Gloaguen, V., et al., Carbohydr Res. 2004 Jan 2;339(l):97-103).
  • Tray dryers accept moist solid on trays. Hot air (or nitrogen) is circulated to dry. Shelf dryers can also employ reduced pressure or vacuum to dry at room temperature when products are temperature sensitive and are similar to a freeze-drier but less costly to use and can be easily scaled-up. Spray dryers are relatively simple in operation, which accept feed in fluid state and convert it into a dried particulate form by spraying the fluid into a hot drying medium.
  • Rotary dryers operate by continuously feeding wet material, which is dried by contact with heated air, while being transported along the interior of a rotating cylinder, with the rotating shell acting as the conveying device and stirrer.
  • Spin flash dryers are used for drying of wet cake, slurry, or paste which is normally difficult to dry in other dryers.
  • the material is fed by a screw feeder through a variable speed drive into the vertical drying chamber where it is heated by air and at the same time disintegrated by a specially designed disintegrator.
  • the heating of air may be direct or indirect depending upon the application.
  • the dry powder is collected through a cyclone separator/bag filter or with a combination of both.
  • Intracellular polysaccharides and cell wall polysaccharides can be purified from whole cell mass (see form example U.S. Patent 4,992,540; U.S. Patent 4,810,646; J Sietsma JH., et al., Gen Microbiol. 1981 Jul;125(l):209-12; Fleet GH, Manners DJ., J Gen Microbiol. 1976 May;94(l): 180-92).
  • a pressure disrupter pumps of a slurry through a restricted orifice valve High pressure (up to 1500 bar) is applied, followed by an instant expansion through an exiting nozzle.
  • Cell disruption is accomplished by three different mechanisms: impingement on the valve, high liquid shear in the orifice, and sudden pressure drop upon discharge, causing an explosion of the cell.
  • the method is applied mainly for the release of intracellular molecules.
  • R is the amount of soluble protein
  • Rm is the maximum amount of soluble protein
  • K is the temperature dependent rate constant
  • N is the number of passes through the homogenizer (which represents the residence time).
  • P is the operating pressure.
  • cell containing samples are cryogenically ball milled in a planetary ball mill (Retsch, PMlOO) at 10-80 g batch size.
  • the powder is placed in a grinding bowl with eight to ten %-inch-diameter stainless steel balls.
  • the sample is cooled repeatedly with liquid nitrogen.
  • the material is milled at 400- 550 rpm for 30 to 60 min.
  • the final product is dried in a desiccator overnight.
  • Another widely applied method is the cell lysis with high frequency sound that is produced electronically and transported through a metallic tip to an appropriately concentrated cellular suspension, ie: sonication.
  • the concept of ultrasonic disruption is based on the creation of cavities in cell suspension.
  • the invention comprises polysaccharides from a species of the genus Porphyridium, optionally sulfated at a level of 4.7 or higher, that have been reduced in molecular weight through milling or shearing such as, for example, sonication or ball milling.
  • Blending high speed or Waring
  • the french press or even centrifugation in case of weak cell walls, also disrupt the cells by using the same concepts.
  • Cells can also be ground after drying in devices such as a colloid mill.
  • the invention includes a method of preparing a dry homogenate by use of lyophilization.
  • a cell containing material is lyophilized prior to homogenization, such as by grinding or milling.
  • the cell containing material comprises both cells (biomass) and soluble polysaccharides in the culture medium. In other embodiments, only biomass is used.
  • the cell-containing material is optionally dried (at least partially) or otherwise concentrated before subsequent lyophilization.
  • the lyophilized material may be homogenized to produce a homogenate comprising a powder or particles of a subcellular size range.
  • Non-limiting examples include particles that are less than 800 microns in diameter, less than 200 microns in diameter, less than 100 microns in diameter, less than 50 microns in diameter, less than 10 microns in diameter, less than 5 microns in diameter, and less than 1 micron in diameter.
  • the average particle size of a composition disclosed herein is less than 800 microns in diameter, less than 200 microns in diameter, less than 100 microns in diameter, less than 50 microns in diameter, less than 10 microns in diameter, less than 5 microns in diameter, and less than 1 micron in diameter.
  • homogenized cells or biomass
  • the homogenized material may improve exposure of the polysaccharides to solvent and subsequent uptake in the digestive tract of a mammal.
  • the homogenization process may fragment the polysaccharides under certain conditions, such as to produce fragments of sulfated polysaccharides, which increase the total activity available in the homogentate.
  • the homogenization process releases intracellular polysaccharides which add to the total activity in the homogenate. While none of these proposed mechanisms may be responsible for the improved activity seen with homogenates of cells, a combination of two or more of the above mechanisms also may be responsible for the improved activity.
  • a biomass homogenate at a 5% or 10% diet reduced TSC to a much greater extent (approximately 39% and 54% respectively) than non- homogenized biomass administered at much higher dosages as discussed in the literature (22% reduction in TSC observed for a 19% diet of unpurified biomass- see Dvir et al. Br. J. Nutri. (2000) 84:469-476). As such it was a surprising result to see that lower dosages of a biomass homogenate generate such a significant reduction in TSC.
  • microalgae cell homogenates can be considered partially pure polysaccharide compositions.
  • Cell disruption aids in increasing the amount of solvent-accessible polysaccharide by breaking apart cell walls that are largely composed of polysaccharide.
  • Homogenizatio ⁇ as described herein can increase the amount of solvent-available polysaccharide significantly.
  • homogenization can increase the amount of solvent-available polysaccharide by at least a factor of 0.25, at least a factor, of 0.5, at least a factor of 1, at least a factor of 2, at least a factor of 3, at least a factor of 4, at least a factor of 5, at least a factor of 8, at least a factor of 10, at least a factor of 15, at least a factor of 20, at least a factor of 25, and at least a factor of 30 or more compared to the amount of solvent- available polysaccharide in an identical or similar quantity of non-homogenized cells of the same type.
  • One way of determining a quantity of cells sufficient to generate a given quantity of homogenate is to measure the amount of a compound in the homogenate and calculate the gram quantity of cells required to generate this amount of the compound using known data for the amount of the compound per gram mass of cells.
  • the quantity of many such compounds per gram of particular microalgae cells are known. For examples, see Figure 9. Given a certain quantity of a compound in a composition, the skilled artisan can determine the number of grams of intact cells necessary to generate the observed amount of the compound.
  • the number of grams of microalgae cells present in the composition can then be used to determine if the composition contains at least a certain amount of solvent- available polysaccharide sufficient to indicate whether or not the composition contains homogenized cells, such as for example five times the amount of solvent-available polysaccharide present in a similar or identical quantity of unhomogenized cells.
  • Assays for detecting polysaccharides can be used to quantitate starting polysaccharide concentration, measure yield during purification, calculate density of secreted polysaccharide in a photobioreactor, measure polysaccharide concentration in a finished product, and other purposes..
  • the phenol: sulfuric acid assay detects carbohydrates (see Hellebust, Handbook of Phycological Methods, Cambridge University Press, 1978; and Cuesta G., et al., J Microbiol Methods. 2003 Jan;52(l):69-73).
  • the 1,6 dimethylmethylene blue assay detects anionic polysaccharides, (see for example Braz J Med Biol Res. 1999 May;32(5):545-50; Clin Chem. 1986 Nov;32(ll):2073-6).
  • Polysaccharides can also be analyzed through methods such as HPLC, size exclusion chromatography, and anion exchange chromatography (see for example Prosky L, Asp N, Schweizer TF, DeVries JW & Furda I (1988) Determination of insoluble, soluble and total dietary fiber in food and food products: Interlaboratory study. Journal of the Association of Official Analytical Chemists 71, 1017 ⁇ 1023; Int J Biol Macromol. 2003 Nov;33(l-3):9-18)
  • Polysaccharides can also be detected using gel electrophoresis (see for example Anal Biochem. 2003 Oct 15;321(2):174-82; Anal Biochem. 2002 Jan l;300(l):53-68).
  • Monosaccharide analysis of polysaccharides can be performed by combined gas chromatography/mass spectrometry (GC/MS) of the per-O-trimethylsilyl (TMS) derivatives of the monosaccharide methyl glycosides produced from the sample by acidic methanolysis (see Merkle and Poppe (1994) Methods Enzymol. 230: 1-15; York, et al. (1985) Methods Enzymol. 118:3-40).
  • GC/MS gas chromatography/mass spectrometry
  • compositions of the invention include a microalgal polysaccharide or homogenate as described herein.
  • the composition may comprise a homogenous or a heterogeneous population of polysaccharide molecules, including sulfated polysaccharides as non-limiting embodiments.
  • homogenous populations include those containing a single type of polysaccharide molecule, such as that with the same structure and molecular weight.
  • Non- limiting examples of heterogeneous populations include those containing more than one type of polysaccharide molecule, such as a mixture of polysaccharides having a molecular weight (MW) within a range or a MW above or below a MW value.
  • MW molecular weight
  • the Porphyridium sp. exopolysaccharide is typically produced in a range of sizes from 3-5 million Daltons.
  • a polysaccharide containing composition of the invention may be optionally protease treated, or reduced in the amount of protein, as described above.
  • a composition of the invention may comprise one or more polysaccharides produced by microalgae that have not been recombinantly modified.
  • the microalgae may be those which are naturally occurring or those which have been maintained in culture in the absence of alteration by recombinant DNA techniques or genetic engineering.
  • the polysaccharides are those from modified microalgae, such as, but not limited to, microalgae modified by recombinant techniques.
  • modified microalgae such as, but not limited to, microalgae modified by recombinant techniques.
  • Non-limiting examples of such techniques include introduction and/or expression of an exogenous nucleic acid sequence encoding a gene product; genetic manipulation to decrease or inhibit expression of an endogenous microalgal gene product; and/or genetic manipulation to increase expression of an endogenous microalgal gene product.
  • the invention contemplates recombinant modification of the various microalgae species described herein.
  • the microalgae is from the genus Porphyridium.
  • Polysaccharides provided by the invention that are produced by genetically modified microalgae or microalgae that are provided with an exogenous carbon source can be distinct from those produced by microalgae cultured in minimal growth media under photoautotrophic conditions (ie: in the absence of a fixed carbon source) at least in that they contain a different monosaccharide content relative to polysaccharides from unmodified microalgae or microalgae cultured in minimal growth media under photoautotrophic conditions.
  • Non-limiting examples include polysaccharides having an increased amount of arabinose (Ara), rhamnose (Rha), fucose (Fuc), xylose (XyI), glucuronic acid (GIcA), galacturonic acid (GaIA), mannose (Man), galactose (Gal), glucose (GIc), N-acetyl galactosamine (GaINAc), N-acetyl glucosamine (GIcNAc), and/or N-acety] neuraminic acid (NANA) 5 per unit mass (or per mole) of polysaccharide, relative to polysaccharides from either non-genetically modified microalgae or microalgae cultured photoautotrophically.
  • An increased amount of a monosaccharide may also be expressed in terms of an increase relative to other monosaccharides rather than relative to the unit mass, or mole, of polysaccharide.
  • An example of genetic modification leading to production of modified polysaccharides is transforming a microalgae with a carbohydrate transporter gene, and culruring a transformant in the presence of a monosaccharide which is transported into the cell from the culture media by the carbohydrate transporter protein encoded by the carbohydrate transporter gene. In some instances the culture can be in the dark, where the monosaccharide, such as glucose, is used as the sole energy source for the cell.
  • the culture is in the light, where the cells undergo photosynthesis and therefore produce monosaccharides such as glucose in the chloroplast and transport the monosaccharides into the cytoplasm, while additional exogenously provided monosaccharides are transported into the cell by the carbohydrate transporter protein.
  • monosaccharides from the cytoplasm are transported into the endoplasmic reticulum, where polysaccharide synthesis occurs.
  • Novel polysaccharides produced by non- genetically engineered microalgae can also be generated by nutritional manipulation, ie: exogenously providing carbohydrates in the culture media that are taken up through endogenous transport mechanisms.
  • Uptake of the exogenously provided carbohydrates can be induced, for example, by culruring the cells in the dark, thereby forcing the cells to utilize the exogenously provided carbon source.
  • Porphyridium cells cultured in the presence of 7% glycerol in the dark produce a novel polysaccharide because the intracellular carbon flux under these nutritionally manipulated conditions is different from that under photosynthetic conditions. Insertion of carbohydrate transporter genes into microalgae facilitates, but is not strictly necessary for, polysaccharide structure manipulation because expression of such genes can significantly increase the concentration of a particular monosaccharide in the cytoplasm of the cell.
  • carbohydrate transporter genes encode proteins that transport more than one monosaccharide, albeit with different affinities for different monosaccharides (see for example Biochimica et Biophysica Acta 1465 (2000) 263-274).
  • a microalgae species can be transformed with a carbohydrate transporter gene and placed under different nutritional conditions, wherein one set of conditions includes the presence of exogenously provided galactose, and the other set of conditions includes the presence of exogenously provided xylose, and the transgenic species produces structurally distinct polysaccharides under the two conditions.
  • the invention provides novel polysaccharides.
  • Nutritional manipulation can also be performed, for example, by culturing the microalgae in the presence of high amounts of sulfate, as described herein.
  • nutritional manipulation includes addition of one or more exogenously provided carbon sources as well as one or more other non-carbohydrate culture component, such as 5OmM MgSO 4 .
  • the increase in one or more of the above listed monosaccharides in a polysaccharide may be from below to above detectable levels and/or by at least about 5%, to at least about 2000%, relative to a polysaccharide produced from the same microalgae in the absence of genetic or nutritional manipulation.
  • the presence of the monosaccharide in a microalgal polysaccharide indicates the presence of a polysaccharide distinct from that in unmodified microalgae.
  • the invention includes modification of these microalgae to incorporate arabinose and/or fucose, because polysaccharides from two strains of these species do not contain detectable amounts of these monosaccharides (see Example 5 herein), hi another non-limiting example, the modification of Porphyridium sp.
  • the invention includes a method of producing a polysaccharide comprising culturing a microalgae cell in the presence of at least about 0.01 micromolar of an exogenously provided fixed carbon compound, wherein the compound is incorporated into the polysaccharide produced by the cell.
  • the compound is selected from Table 2 or 3.
  • the cells may optionally be selected from the species listed in Table 1, and cultured by modification, using the methods disclosed herein, or the culture conditions also lusted in Table 1.
  • the methods may also be considered a method of producing a glycopolymer by culturing a transgenic microalgal cell in the presence of at least one monosaccharide, wherein the monosaccharide is transported by the transporter into the cell and is incorporated into a microalgal polysaccharide.
  • the cell is selected from Table 1, such as where the cell is of the genus Porphyridium, as a non-limiting example. In some cases, the cell is selected from Porphyridium sp. and Porphyridium cruentum.
  • Embodiments include those wherein the polysaccharide is enriched for the at least one monosaccharide compared to an endogenous polysaccharide produced by a non-transgenic cell of the same species.
  • the monosaccharide may be selected from Arabinose, Fructose, Galactose, Glucose, Mannose, Xylose, Glucuronic acid, Glucosamine, Galactosamine, Rhamnose and N-acetyl glucosamine.
  • transgenic cell expressing a sugar transporter
  • the transporter has a lower K n , for glucose than at least one monosaccharide selected from the group consisting of galactose, xylose, glucuronic acid, mannose, and rhamnose.
  • the transporter has a lower K m for galactose than at least one monosaccharide selected from the group consisting of glucose, xylose, glucuronic acid, mannose, and rhamnose.
  • the transporter has a lower K m for xylose than at least one monosaccharide selected from the group consisting of glucose, galactose, glucuronic acid, mannose, and rhamnose. In further embodiments, the transporter has a lower K m for glucuronic acid than at least one monosaccharide selected from the group consisting of glucose, galactose, xylose, mannose, and rhamnose. In yet additional embodiments, the transporter has a lower K m for mannose than at least one monosaccharide selected from the group consisting of glucose, galactose, xylose, glucuronic acid, and rhamnose.
  • the transporter has a lower K m for rhamnose than at least one monosaccharide selected from the group consisting of glucose, galactose, xylose, glucuronic acid, and mannose.
  • These general methods can also be used in cells other than microalgae, for example, bacteria that produce polysaccharides.
  • the cell is cultured in the presence of at least two monosaccharides, both of which are transporter by the transporter.
  • the two monosaccharides are any two selected from glucose, galactose, xylose, glucuronic acid, rhamnose and mannose.
  • the method comprises providing a transgenic cell containing a recombinant gene encoding a monosaccharide transporter; and culturing the cell in the presence of at least one monosaccharide, wherein the monosaccharide is transported by the transporter into the cell and is incorporated into a polysaccharide of the cell.
  • transportation of a monosaccharide from the media into a microalgal cell allows for the monosaccharide to be used as an energy source, as disclosed below, and for the monosaccharide to be transported into the endoplasmic reticulum (ER) by cellular transporters.
  • ER endoplasmic reticulum
  • polysaccharide production and glycosylation occurs such that in the presence of exogenously provided monosaccharides, the sugar content of the microalgal polysaccharides change.
  • the invention includes a novel microalgal polysaccharide, such as from microalgae of the genus Porphyridium, comprising detectable amounts of xylose, glucose, and galactose wherein the molar amount of one or more of these three monosaccharides in the polysaccharide is not present in a polysaccharide of Porphyridium that is not genetically or nutritionally modified.
  • a non-nutritionally and non- genetically modified Porphyridium polysaccharide can be found, for example, in Jones R., Journal of Cellular Comparative Physiology 60; 61-64 (1962).
  • the amount of glucose, in the polysaccharide is at least about 65% of the molar amount of galactose in the same polysaccharide.
  • glucose is at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 100%, at least about 105%, at least about 110%, at least about 120%, at least about 130%, at least about 140%, at least about 150%, at least about 200%, at least about 250%, at least about 300%, at least about 350%, at least about 400%, at least about 450%, at least about 500%, or more, of the molar amount of galactose in the polysaccharide.
  • Further embodiments of the invention include those wherein the amount of glucose in a microalgal polysaccharide is equal to, or approximately equal to, the amount of galactose (such that the amount of glucose is about 100% of the amount of galactose). Moreover, the invention includes microalgal polysaccharides wherein the amount of glucose is more than the amount of galactose.
  • the amount of glucose, in the polysaccharide is less than about 65% of the molar amount of galactose in the same polysaccharide.
  • the invention thus provides for polysaccharides wherein the amount of glucose is less than about 60%, less than about 55%, less than about 50%, less than about 45%, less than about 40%, less than about 35%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, or less than about 5% of the molar amount of galactose in the polysaccharide.
  • the invention includes a microalgal polysaccharide, such as from microalgae of the genus Porphyridium, comprising detectable amounts of xylose, glucose, galactose, mannose, and rhamnose, wherein the molar amount of one or more of these five monosaccharides in the polysaccharide is not present in a polysaccharide of non-genetically modified and/or non-nutritionally modified microalgae.
  • the amount of rhamnose in the polysaccharide is at least about 100% of the molar amount of mannose in the same polysaccharide.
  • rhamnose is at least about 110%, at least about 120%, at least about 130%, at least about 140%, at least about 150%, at least about 200%, at least about 250%, at least about 300%, at least about 350%, at least about 400%, at least about 450%, or at least about 500%, or more, of the molar amount of mannose in the polysaccharide.
  • Further embodiments of the invention include those wherein the amount of rhamnose in a microalgal polysaccharide is more than the amount of mannose on a molar basis.
  • the amount of rhamnose, in the polysaccharide is less than about 75% of the molar amount of mannose in the same polysaccharide.
  • the invention thus provides for polysaccharides wherein the amount of rhamnose is less than about 70%, less than about 65%, less than about 60%, less than about 55%, less than about 50%, less than about 45%, less than about 40%, less than about 35%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, or less than about 5% of the molar amount of mannose in the polysaccharide.
  • the invention includes a microalgal polysaccharide, such as from microalgae of the genus Porphyridium, comprising detectable amounts of xylose, glucose, galactose, mannose, and rhamnose, wherein the amount of mannose, in the polysaccharide, is at least about 130% of the molar amount of rhamnose in the same polysaccharide.
  • mannose is at least about 140%, at least about 150%, at least about 200%, at least about 250%, at least about 300%, at least about 350%, at least about 400%, at least about 450%, or at least about 500%, or more, of the molar amount of rhamnose in the polysaccharide.
  • the amount of mannose, in the polysaccharide is equal to or less than the molar amount of rhamnose in the same polysaccharide.
  • the invention thus provides for polysaccharides wherein the amount of mannose is less than about 95%, less than about 90%, less than about 85%, less than about 80%, less than about 75%, less than about 70%, less than about 65%, less than about 60%, less than about 60%, less than about 55%, less than about 50%, less than about 45%, less than about 40%, less than about 35%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, or less than about 5% of the molar amount of rhamnose in the polysaccharide.
  • the invention includes a microalgal polysaccharide, such as from microalgae of the genus Porphyridium, comprising detectable amounts of xylose, glucose, and galactose, wherein the amount of galactose in the polysaccharide, is at least about 100% of the molar amount of xylose in the same polysaccharide.
  • a microalgal polysaccharide such as from microalgae of the genus Porphyridium, comprising detectable amounts of xylose, glucose, and galactose, wherein the amount of galactose in the polysaccharide, is at least about 100% of the molar amount of xylose in the same polysaccharide.
  • rhamnose is at least about 105%, at least about 110%, at least about 120%, at least about 130%, at least about 140%, at least about 150%, at least about 200%, at least about 250%, at least about 300%, at least about 350%, at least about 400%, at least about 450%, or at least about 500%, or more, of the molar amount of mannose in the polysaccharide.
  • Further embodiments of the invention include those wherein the amount of galactose in a microalgal polysaccharide is more than the amount of xylose on a molar basis.
  • the amount of galactose, in the polysaccharide is less than about 55% of the molar amount of xylose in the same polysaccharide.
  • the invention thus provides for polysaccharides wherein the amount of galactose is less than about 50%, less than about 45%, less than about 40%, less than about 35%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, or less than about 5% of the molar amount of xylose in the polysaccharide.
  • the invention includes a microalgal polysaccharide, such as from microalgae of the genus Porphyridium, comprising detectable amounts of xylose, glucose, glucuronic acid and galactose, wherein the molar amount of one or more of these five monosaccharides in the polysaccharide is not present in a polysaccharide of unmodified microalgae.
  • the amount of glucuronic acid, in the polysaccharide is at least about 50% of the molar amount of glucose in the same polysaccharide.
  • glucuronic acid is at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 100%, at least about 105%, at least about 110%, at least about 120%, at least about 130%, at least about 140%, at least about 150%, at least about 200%, at least about 250%, at least about 300%, at least about 350%, at least about 400%, at least about 450%, or at least about 500%, or more, of the molar amount of glucose in the polysaccharide.
  • Further embodiments of the invention include those wherein the amount of glucuronic acid in a microalgal polysaccharide is more than the amount of glucose on a molar basis.
  • the exopolysaccharide, or cell homogenate polysaccharide comprises glucose and galactose wherein the molar amount of glucose in the exopolysaccharide, or cell homogenate polysaccharide, is at least about 55% of the molar amount of galactose in the exopolysaccharide or polysaccharide.
  • the molar amount of glucose in the exopolysaccharide, or cell homogenate polysaccharide is at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, or at least about 100% of the molar amount of galactose in the exopolysaccharide or polysaccharide.
  • the invention includes a microalgal polysaccharide, such as from microalgae of the genus Porphyridium, comprising detectable amounts of xylose, glucose, glucuronic acid, galactose, at least one monosaccharide selected from arabinose, fucose, N-acetyl galactosamine, and N-acetyl neuraminic acid, or any combination of two or more of these four monosaccharides.
  • a microalgal polysaccharide such as from microalgae of the genus Porphyridium, comprising detectable amounts of xylose, glucose, glucuronic acid, galactose, at least one monosaccharide selected from arabinose, fucose, N-acetyl galactosamine, and N-acetyl neuraminic acid, or any combination of two or more of these four monosaccharides.
  • Embodiments of the invention include a composition comprising a intact red microalgae cells or a homogenate of red microalgae cells.
  • Non-limiting examples include a powder or particles as disclosed herein.
  • the composition further includes a carrier suitable for consumption by a mammal, such as a human. Therefore, embodiments of the invention include a food product, such as a human food product, comprising a homogenate of red microalgae cells.
  • the composition may be a capsule or tablet comprising the homogenate.
  • compositions wherein the red microalgae cells are of the genus Porphyridium.
  • Non-limiting examples include red microalgae cells that are Porphyridium sp. UTEX 637, or a strain derived from Porphyridium sp. UTEX 637; Porphyridium cruentum UTEX 161, or a strain derived from Porphyridium cruentum UTEX 161; Porphyridium aerugineum, or a strain derived from Porphyridium aerugineum; Porphyridium sordidum, or a strain derived from Porphyridium sordidum; or Porphyridium purpureum, or a strain derived from Porphyridium purpureum.
  • compositions wherein the homogenate comprises a sulfated exopolysaccharide, secreted by the red microalgae cells, wherein the polysaccharide in purified form contains at least about 4.6% sulfur, at least about 4.75% sulfur, at least about 5.0% sulfur, at least about 5.25% sulfur, at least about 5.5% sulfur, at least about 5.75% sulfur, at least about 6.0% sulfur, at least about 6.25% sulfur, at least about 6.5% sulfur, at least about 6.75% sulfur, or at least about 7.0% sulfur by weight.
  • compositions wherein the homogenate comprises at least about two, at least about three, at least about four, at least about five, at least about six, at least about seven, at least about eight, at least about nine, at least about 10, at least about 12, at least about 14, at least about 16, at least about 18, or at least about 20 times the amount of solvent-available polysaccharide compared to the quantity of unhomogenized cells (biomass) used to form the homogenate.
  • the homogenate contains from about two, to about 20 or more, times the amount or level of solvent-available polysaccharide compared to the unhomogenized cells needed to generate the microalgal cell homogenate.
  • Embodiments of the invention also include compositions comprising combinations of the above.
  • the composition comprises a homogenate with a sulfated exopolysaccharide secreted by red microalgae cells that contains at least 3.0%, or at least 4.75%, sulfur by weight, and the homogenate contains at least two times the amount of solvent-available polysaccharide as present in a quantity of unhomogenized cells needed to generate the microalgal cell homogenate.
  • the invention includes mixtures of polysaccharides from Porphyridium as described herein and polysaccharides from a species of Chlorella, such as Chlorella marina, Chlorella salina, Chlorella pyrenoidosa, Chlorella vulgaris, Chlorella anitrata, Chlorella antarctica, Chlorella autotrophica, Chlorella regularis, among others (see World Catalog of Algae, 2.sup.nd Edition, pages 58-74; Miyachi et al.
  • Chlorella marina Chlorella marina
  • Chlorella salina Chlorella pyrenoidosa
  • Chlorella vulgaris Chlorella anitrata
  • Chlorella antarctica Chlorella autotrophica
  • Chlorella regularis among others (see World Catalog of Algae, 2.sup.nd Edition, pages 58-74; Miyachi et al.
  • Polysaccharides from microalgae can be formulated for ingestion to achieve a hypocholesterolemic effect.
  • the secreted polysaccharide from Porphyridium sp. can be formulated for administration as a cholesterol lowering agent.
  • Secreted polysaccharides from Porphyridium cruentum, Porphyridium purpureum, Porphyridium aerugineum, Rhodella maculata, Rhodella reticulata, Chlorella autotrophica, Chlorella stigmatophora, Chlorella capsulata, Achnanthes brevipes and Achnanthes longipes can also be formulated for administration as a cholesterol lowering agent.
  • microalgae are cultured, for example, in photobioreactors in the presence of light, more preferably in the presence of strong light such as 175 ⁇ mol photons per square meter per second, for a period of time sufficient for the cells to secrete polysaccharide molecules.
  • Some species such as those of Chlorella and Porphyridium, can also be cultured in the absence of light and in the presence of a fixed carbon source.
  • the polysaccharides or polysaccharide material will be from a Porphyridium species, such as one that has been subject to genetic and/or nutritional manipulation to produce polysaccharides with altered monosaccharide content and/or altered sulfation.
  • Patients in need of cholesterol lowering polysaccharide agents such as polysaccharides are preferably those with total cholesterol above 200 mg/dL, those with LDL Cholesterol above 130 mg/dL, those with HDL Cholesterol less than 40 mg/dL, and those with triglycerides above 150 mg/dL.
  • the invention also comprises administering to a patient described herein a combination of an algal polysaccharide such as that from a cell of the genus Porphyridium and another compound such as a plant phytosterol or a statin such as Pravachol ® , Mevacor ® , Zocor ® , Lescol ® , Lipitor ® , Baycol ® , Crestor ® , and Advicor ® .
  • the invention also comprises a method of reducing the side effects of a statin drug comprising lowering the dosage of a statin and administering a polysaccharide produced from microalgae, such as for example the polysaccharide from a cell of the genus Porphyridium.
  • statins include nausea, irritability and short temper, hostility, homicidal impulses, loss of mental clarity, amnesia, kidney failure, diarrhea, muscle aching and weakness, tingling or cramping in the legs, inability to walk, sleeping problems, constipation, impaired muscle formation, erectile dysfunction, temperature regulation problems, nerve damage, mental confusion, liver damage and abnormalities, neuropathy, and destruction of CoQlO.
  • the invention also includes administering a polysaccharide produced from microalgae, such as for example the polysaccharide from a cell of the genus Porphyridium, to a patient with total cholesterol of 240 mg/dL or more; to a patient with LDL Cholesterol of 130 to 159 mg/dL, 160 to 189 mg/dL, and 190 mg/dL or higher; and to a patient with triglycerides of 150 to 199 mg/dL, or 200 mg/dL or higher.
  • a polysaccharide produced from microalgae such as for example the polysaccharide from a cell of the genus Porphyridium
  • cells of the genus Porphyridium are harvested from culture and homogenized to form a composition for administration to lower cholesterol. Homogenization of the cells provides an increased level of bioavailability of the cell wall polysaccharide compared to intact cells. Homogenization can be performed by methods such as sonication, jet milling, colloid milling, wet grinding, dry grinding, and other methods.
  • a preferred composition for cholesterol reduction is homogenized Porphyridium, wherein the average particle size is less then 300 microns, more preferably less than 200 microns, more preferably less than 100 microns, more preferably less than 50 microns, more preferably less than 25 microns, and more preferably less than 10 microns.
  • the cells are dried before grinding, while in other embodiments homogenization is performed on wet cells, such as sonication.
  • Homogenization of microalgae to increase bioavailability of cell wall polysaccharides can be performed to produce homogenates, also referred to herein as polysaccharide material, of any microalgae, including species from Table 1.
  • Polysaccharides of the invention may be formulated as a composition for oral consumption, as in a dietary supplement as a non- limiting example.
  • the formulation may be in solid or liquid form.
  • purified lyophilized polysaccharide can be formulated in capsules or tablets. Conventional methods for the preparation of capsules or tablets are known to the skilled person. The methods may include use of pharmaceutically .
  • acceptable excipients such as binding agents, fillers, disintegrants, or wetting agents, sweeteners, including, pregelatinised maize starch, polyvinylpyrrolidone, hydroxypropyl methylcellulose, fillers, lactose, microcrystalline cellulose, calcium hydrogen phosphate, lubricants, magnesium stearate, talc, silica, potato starch or sodium starch glycolate, sodium lauryl sulfate, mannitol, lactose, starch, magnesium stearate, polyvinyl pyroUidone, sodium saccharine, cellulose and magnesium carbonate in the formation of a capsule or tablet.
  • sweeteners including, pregelatinised maize starch, polyvinylpyrrolidone, hydroxypropyl methylcellulose, fillers, lactose, microcrystalline cellulose, calcium hydrogen phosphate, lubricants, magnesium stearate, talc, silica, potato starch or sodium starch glycolate, sodium lauryl
  • the capsule may be comprise a slow-dissolving polymers.
  • Non-limiting polymers include sodium carboxymethylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose and hydroxyethylcellulose.
  • Other preferred cellulose ethers are known to the skilled person (Alderman, Int. J. Pharm. Tech. & Prod. Mfr., 1984, 5(3): 1-9).
  • the polysaccharide material can be directly encapsulated within a capsule or formed into microspheres that are encapsulated. The formation of microspheres may be by a variety of methods known to the skilled person.
  • the polysaccharide(s) are dispersed in a liquid form, such as in an aqueous solution.
  • the liquid is sprayed onto a core particle, such as a nonpareil composed of sugar and/or starch. This forms a microsphere, which may then be dried, or otherwise processed, before being packaged into capsules.
  • the polysaccharide material can be formed into a solid tablet, optionally with one or more of the excipients listed above.
  • a tablet may be coated by methods known to the skilled person.
  • Solid oral administration can be formulated to give controlled release of the polysaccharide material.
  • Polysaccharide material may also be formulated into capsule form as a liquid.
  • the liquid may be any suitably formulated for inclusion in a capsule as known to the skilled person.
  • the liquid is suitably viscous and does not solvate the capsule to result in leakage from the capsule.
  • the liquid may be a preparation that is a variation of those used in other oral administration, such as those in the form of solutions, syrups, or suspensions, all of which may also be used in the practice of the invention.
  • Such liquid preparations can be prepared by conventional means known to the skilled person with pharmaceutically acceptable additives such as, but not limited to, suspending agents, e.g., sorbitol syrup, cellulose derivatives, or hydrogenated edible fats; emulsifying agents, e.g., lecithin or acacia; non-aqueous vehicles, e.g., almond oil, oily esters, ethyl alcohol, or fractionated vegetable oils; and preservatives, e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid.
  • the preparations can also contain buffer salts, flavoring, coloring, and/or sweetening agents as appropriate.
  • polysaccharide material can be formulated as a food additive.
  • dried polysaccharide can be resuspended in a food substance such as a salad dressing or another sauce or condiment.
  • the material can be formulated into a processed food item.
  • Non-limiting examples include dried foods, canned foods, bars, and frozen foods.
  • Dried foods include dehydrated foods (which are normally rehydrated before consumption), dry cereals, and crackers as non-limiting examples.
  • the polysaccharide material can be formulated into a liquid preparation and for administration as a beverage.
  • a beverage can be alcoholic, nonalcoholic beverage, carbonated, or a health beverage.
  • Such beverage may comprise one or more of the polysaccharides and/or homogenates described herein as well as, optionally, any one or more of the following: a vitamin, electrolyte substitute, caffeine, an amino acid, minerals, artificial and/or natural sweeteners, milk or dry-milk powder, plant phytosterols, and other additives and preserving agents.
  • Additional carriers of the invention include but are not limited to water, salt solutions (e.g., NaCl), saline, buffered saline, alcohols, glycerol, ethanol, gum arabic, vegetable oils, benzyl alcohols, polyethylene glycols, gelatin, carbohydrates such as lactose, amylose or starch, dextrose, magnesium stearate, talc, silicic acid, viscous paraffin, perfume oil, fatty acid esters, hydroxymethylcellulose, polyvinyl pyrolidone, etc., as well as any two or more of the foregoing in combination.
  • the solid or liquid compositions described herein may be advantageously used as a cholesterol lowering composition.
  • a composition may comprise 1) a purified microalgal exopolysaccharide or amicroalgal cell homogenate (ie: polysaccharide material) and 2) a carrier suitable for human oral consumption as described.
  • the exopolysaccharide or cell homogenate may be produced from cells of the genus Porphyridium as a non-limiting example. As disclosed herein, the exopolysaccharide may be substantially free of protein.
  • embodiments of the invention include a composition comprising a homogenate of red microalgae cells, such as cells of the genus Porphyridium, and at least one additional cholesterol lowering agent or compound.
  • the agent or compound is selected from a plant phytosterol, a limonoid, a flavonoid, a tocotrienol, or a combination thereof.
  • the agent is an inhibitor of HMG CoA reductase, such as a statin.
  • the weight/weight ratio of cell material (such as homogenate) to plant phytosterol may range from at least about 0.5:1 to at least about 100:1. Additional non-limiting examples include weight/weight ratios of cell material (such as homogenate) to plant phytosterol of at least about 10:1, at least about 20:1, at least about 30:1, at least about 40:1, at least about 50:1, at least about 60:1, at least about 70:1, at least about 80:1, or at least about 90:1.
  • phytosterols are steroids derived from a plant, yeast or fungus. Phytosterols have a hydroxyl group at C-3 and no other functional groups. Plant and animal sterols differ.
  • the term phytosterol and/or phytostanol according to the invention includes analogues of phytosterols, such as those with a double bond at the 5-position in the cyclic unit as in most natural phytosterols, or one or more double bonds at other positions (e.g. 6, 7, 8(9), 8(14), 14, 5/7, or no double bonds in the cyclic unit as in the stands, or even additional methyl groups as e.g. in alpha- 1 -sitosterol.
  • the invention also includes the use of a mixture of phytosterol and phytostanol with red microalgae cell material.
  • phytosterol and/or phytostanols are those obtained from vegetable oil or wood pulp.
  • Non-limiting examples include alpha-sitosterol, beta- sitosterol, gamma-sitosterol, stigmasterol, ergosterol, campesterol, avenasterol, desmosterol, chalinosterol, poriferasterol, clionasterol, sitostanol, alpha-sitostanol, beta-sitostanol, stigmastanol, campestanol or brassicasterol.
  • at least 44 phytosterols have been identified and they may be used in the practice of the invention as deemed appropriate and/or desirable by the skilled person.
  • oryzanol may be used.
  • Phytosterols have been reported and are known in the field (see “Advance in Lipid Research", pages 193-218, Paoletti, and Kiritchevsky, (Eds) Academic press, NY, (1993); “Effect of Plant Sterols on Lipids and Atherosclerosis", Pollack, O. J., Pharmac, Ther., 31, 177-208 (1985); Annu Rev Plant Biol. 2004;55:429-57; Metabolism. 2006 Mar;55(3):391 - 5; Am J ClinNutr. 2005 Jun;81 (6): 1351-8; Lipids. 2005 Feb;40(2): 169-74; Lipids.
  • the plant phytosterol is a preparation of wood-derived phytosterol.
  • the wood-derived phytosterol preparation comprises at least about 8% stanol, at least about 40% sitosterol, and at least about 20% campesterol.
  • the wood-derived phytosterol preparation comprises between about 5% and about 20% stanol, between about 40% and about 60% sitosterol, and between about 15% and about 35% campesterol.
  • the weight/weight ratio of cell material (such as homogenate) to limonoid may range from at least about 25:1 to at least about 900:1. Additional non- limiting examples include weight/weight ratios of cell material (such as homogenate) to limonoid of at least about 50:1, at least about 100:1, at least about 200:1, at least about 300:1, at least about 400:1, at least about 500:1, at least about 600:1, at least about 700:1, or at least about 800:1.
  • Limonoid compounds are known to the skilled person, and they include limonin or nomilin, which may be present in a composition as described herein.
  • the weight/weight ratio of cell material (such as homogenate) to flavonoid may range from at least about 40:1 to at least about 800:1. Additional non- limiting examples include weight/weight ratios of cell material (such as homogenate) to flavonoid of at least about 70:1, at least about 100:1, at least about 200:1, at least about 300:1, at least about 400:1, at least about 500:1, at least about 600:1, or at least about 700:1.
  • Flavonoid compounds are known to the skilled person, and they include polymefhoxylated flavones (pmfs), which decrease apoprotein b (a structural protein needed for endogenous synthesis of LDL and cholesterol).
  • pmfs polymefhoxylated flavones
  • apoprotein b a structural protein needed for endogenous synthesis of LDL and cholesterol.
  • the pmfs tangeretin and nobiletin decrease diacylglycerol acetyl transferase (a liver enzyme needed for endogenous synthesis of triglycerides) activity.
  • Some compositions of the invention comprise a flavonoid selected from hesperidin, naringin, naringenin, hesperitin, nobiletin or tangeretin.
  • the weight/weight ratio of cell material (such as homogenate) to flavonoid may range from at least about 30:1 to at least about 900:1. Additional non-limiting examples include weight/weight ratios of cell material (such as homogenate) to flavonoid of at least about 50:1, at least about 100:1, at least about 200:1, at least about 300:1, at least about 400:1, at least about 500:1, at least about 600:1, at least about 700:1, or at least about 800:1.
  • a composition of the invention may comprise one or more tocotrienol, like alpha-tocotrienol, gamma tocotrienol, and delta-tocotrienol as non-limiting examples.
  • compositions comprising a homogenate of red microalgae cells and at least two, or at least three, compounds selected from a plant phytosterol, a limonoid, a flavonoid, and a tocotrienol.
  • the composition comprises a plant phytosterol and at least one flavonoid selected from hesperidin, naringin, naringenin, hesperitin, nobiletin and tangeretin.
  • Additional embodiments of the invention include methods of formulating the above described compositions.
  • the method may comprise combining or mixing the homogenate and the one or more cholesterol lowering agent or compound.
  • the homogenate may be prepared as described herein, such as by culturing cells of the genus Porphyridium; separating the cells from culture media to prepare a cell material (biomass in this instance); drying (at least partially) the cell material; and homogenizing/disrupting the cell material.
  • the cells and polysaccharides in the media are isolated together and then dried or concentrated (at least partially) before homogenization.
  • kits or compositions comprising red microalgae cell material, such as cell material of the genus Porphyridium, and at least one cholesterol lowering component.
  • the combination of cell material and the at least one component is formulated, or packaged, for use or sale as a single unit.
  • the cell material comprises cells per se, while in other embodiments, the cell material is a polysaccharide containing fraction of a preparation of cells (biomass).
  • the cholesterol lowering component may be an agent or compound selected from a plant phytosterol, a limonoid, a flavonoid, a tocotrienol, or a combination thereof as described herein.
  • the agent is an inhibitor of HMG CoA reductase, such as a statin.
  • composition is formulated to comprise the cell material and the at least one component in contact with each other.
  • the cell material is not in contact with the component as these materials are present together in the form of a kit.
  • it may further comprise instructions or a label directing the mixing of the cell material (such as a homogenate) and at least one component in a specific ratio, such as by weight or by mole.
  • the instructions or a label may direct ingestion of the cell material and the at least one component in specific dosages.
  • An additional embodiment of the invention includes instructions or a label directing administration of the cell material and at least one component to lower cholesterol levels in a subject or as part of ongoing treatments or therapies to lower cholesterol levels.
  • a further embodiment of the invention includes instructions or a label directing administration of the cell material and at least one component in a specific order, such as ingestion of the cell material and at least one component over a specified time period or with separation by a specific time period.
  • Further embodiments of the invention include methods of formulating the above described composition comprising cell material and at least one cholesterol lowering agent or compound.
  • the method may comprise combining or mixing cells, optionally dried (at least partially), and the one or more agent or compound.
  • the method comprises culturing cells of the genus Porphyridium; separating the cells from culture media to prepare a cell containing material (biomass); drying (at least partially) the cell containing material; and mixing the dried material with at least one cholesterol lowering agent or compound.
  • Non-limiting examples of drying methods include tray drying, spin drying, rotary drying, spin flash drying, and lyophilization.
  • the cell containing material is homogenized before mixing with the at least one agent or compound to generate a mixture.
  • methods for homogenization include pressure disruption, sonication, jet milling and ball milling.
  • the cell containing material is homogenized such that the homogenate contains at least two times the amount of solvent-available polysaccharide present in a quantity of unhomogenized cells needed to generate the homogenate.
  • the cells are washed before homogenization.
  • Additional embodiments include methods wherein the cells are cultured in at least 100, at least 200, at least 300, at least 400, at least 500, at least 600, or at least 700 mM sulfate.
  • the sulfate may be provided in at least one form selected from Na 2 SO 4 -IO H 2 O, MgSO 4 *7H 2 0, MnSO 4 , and CuSO 4 .
  • the resultant homogenate may contain sulfated exopolysaccharides with at least about 4.6% sulfur by weight of purified polysaccharide, at least about A.I '5% sulfur by weight of purified polysaccharide, at least about 5.0% sulfur by weight of purified polysaccharide, at least about 5.25% sulfur by weight of purified polysaccharide, at least about 5.5% sulfur by weight of purified polysaccharide, at least about 5.75% sulfur by weight of purified polysaccharide, at least about 6.0% sulfur by weight of purified polysaccharide, at least about 6.25% sulfur by weight of purified polysaccharide, at least about 6.5% sulfur by weight of purified polysaccharide, at least about 6.75% sulfur by weight of purified polysaccharide, or at least about 7.0% sulfur by weight by weight of purified polysaccharide.
  • the method of preparing the composition may further comprise adding a carrier suitable for human consumption, or a carrier for oral consumption or administration to a mammal, into the mixture of cell material and at least one cholesterol lowering agent or compound.
  • the cholesterol lowering compositions of the invention may be administered to a subject in need thereof by any appropriate means.
  • Subjects in need of lower cholesterol levels include human beings, who may be tested for serum or plasma cholesterol levels as commonly practiced in clinical medicine by the skilled person. Based on such tests, an elevated cholesterol level in need of lowering may be identified and treated by the methods of the invention.
  • the cholesterol to be lowered is that of low density lipoprotein (LDL) in serum.
  • the cholesterol to be lowered is that of Lp(a), a genetic variation of plasma LDL .
  • the invention includes a method of lowering cholesterol, said method comprising administering a polysaccharide, as disclosed herein, produced by microalgae.
  • the administering is oral, optionally with a biologically acceptable carrier.
  • the polysaccharide is produced by microalgae selected from Table 1. In some embodiments, the polysaccharide is produced by microalgae of the genus Porphyridium.
  • the administered polysaccharide may be a component of a food composition as a non-limiting example. In one range of embodiments, the amount of polysaccharide administered to a human is from about 0.1 to about 50 grams per day. Additional ranges of the invention include an amount of polysaccharide from about 0.25 to about 50 grams per day, about 0.5 to about 5 grams per day, about 0.75 to about 4 grams per day, or about 1 to about 3 grams per day.
  • the polysaccharide is administered via a composition comprising a homogenate of red microalgae cells as described herein.
  • the amount of the composition may be that which contains sufficient homogenate to provide the equivalent amount of polysaccharide as described above.
  • inventions include a method of lowering cholesterol levels in a subject, such as a human patient, by administering a composition comprising a homogenate of red microalgae cells as described herein.
  • the method may comprise oral administration of the composition.
  • the amount of the composition to administer, and be sufficient (or effective) to lower serum cholesterol levels, may be determined by the skilled person by routine and/or repetitive methods.
  • the amount of homogenate administered is in the range of about 0.005 to about 5 grams per kilogram of mammalian body weight per day.
  • amounts to use include about 0.01, about 0.025, about 0.05, about 0.075, about 0.1, about 0.25, about 0.5, about 0.75, about 1, about 1.5, about 2, about 2.5, about 3, about 3.5, about 4, or about 4.5 grams per kg of body weight per day.
  • the composition comprises at least one cholesterol lowering agent or compound, such as, but not limited to, a plant phytosterol, a limonoid, a flavonoid, a tocotrienol, or a combination thereof.
  • the administered amount thereof may be in the range of about 1 to about 500 mg/day, such as about 10, about 20, about 50, about 100, about 150, about 200, about 250, about 300, about 350, about 400, or about 450 mg/day.
  • the administered amount thereof may be in the range of about 200 to about 5000 mg/day, such as about 250, about 300, about 350, about 400, about 450, about 500, about 600, about 700, about 800, about 900, about 1000, about 1500, about 2000, about 2500, about 3000, about 3500, about 4000, or about 4500 mg/day.
  • the administered amount thereof may be in the range of about 1 to about 1200 mg/day, such as about 10, about 20, about 50, about 100, about 150, about 200, about 250, about 300, about 350, about 400, about 450, about 500, about 600, about 700, about 800, about 900, about 1000, or about 1100 mg/day.
  • compositions comprising polysaccharides, whole cell extracts, or mixtures of polysaccharides and whole cell extracts, are provided for topical application or non- systemic administration.
  • the polysaccharide may be any one or more of the microalgal polysaccharides disclosed herein, including those produced by a species, or a combination of two or more species, in Table 1.
  • a whole cell extract may be that prepared from a microalgal species, or a combination of two or more species, in Table 1.
  • polysaccharides, such as exopolysaccharides, and cell extracts from microalgae of the genus Porphyridiwn are used in the practice of the invention.
  • a composition of the invention may comprise from between about 0.001% and about 100%, about 0.01% and about 90%, about 0.1% and about 80%, about 1% and about 70%, about 2% and about 60%, about 4% and about 50%, about 6% and about 40%, about 7% and about 30%, about 8% and about 20%, or about 10% polysaccharide, cell extract, by weight.
  • Topical compositions are usually formulated with a carrier, such as in an ointment or a cream, and may optionally include a fragrance.
  • a carrier such as in an ointment or a cream
  • a fragrance may optionally include a fragrance.
  • topical compositions include gels, solutions, impregnated bandages, liposomes, or biodegradable microcapsules as well as lotions, sprays, aerosols, suspensions, dusting powder, impregnated bandages and dressings, biodegradable polymers, and artificial skin.
  • topical formulations include gels, solutions, impregnated bandages, liposomes, or biodegradable microcapsules as well as lotions, sprays, aerosols, suspensions, dusting powder, impregnated bandages and dressings, biodegradable polymers, and artificial skin.
  • Another non-limiting example of a topical formulation is that of an ophthalmic preparation.
  • Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water.
  • the composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2- octyldodecanol, benzyl alcohol and water.
  • the polysaccharides contain fucose moieties.
  • the polysaccharides are sulfated, such as exopolysaccharides from microalgae of the genus Porphyridium.
  • the polysaccharides will be those from a Porphyridium species, such as one that has been subject to genetic and/or nutritional manipulation to produce polysaccharides with altered monosaccharide content and/or altered sulfation.
  • a composition of the invention comprises a microalgal cell homogenate and a topical carrier.
  • the homogenate may be that of a species listed in Table 1 or may be material produced by a species in the table.
  • a composition comprising purified microalgal polysaccharide and a carrier suitable for topical administration also contains a fusion (or chimeric) protein associated with the polysaccharide.
  • the fusion protein comprises a first protein, or polypeptide region, with at least about 60% amino acid identity with the protein of SEQ ID NO: 28.
  • the first protein has at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 98%, or higher, amino acid identity with the sequence of SEQ ID NO:28.
  • the fusion protein may comprise a second protein, or polypeptide region, with a homogenous or heterologous sequence.
  • the second protein include an antibody and an enzyme.
  • the enzyme is superoxide dismutase, such as that has at least about 60% amino acid identity with the sequence of SEQ ID NO: 14, SEQ ID NO: 15, and a superoxide dismutase selected from Table 16 as non- limiting examples.
  • the superoxide dismutase has at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 98%, or higher, amino acid identity with the sequences of SEQ ID NO: 14 or 15 or from Table 16.
  • the second protein is an antibody.
  • antibodies for use in this aspect of the invention include an antibody that selectively binds to an antigen from a pathogen selected from HIV, Herpes Simplex Virus, gonorrhea, Chlamydia, Human Papillomavirus, and Trichomoniasis.
  • the antibody is a humanized antibody.
  • Polysaccharide compositions for topical application can be formulated by first preparing a purified preparation of polysaccharide.
  • the polysaccharide from aqueous growth media is precipitated with an alcohol, resuspended in a dilute buffer, and mixed with a carrier suitable for application to human skin or mucosal tissue, including the vaginal canal.
  • the polysaccharide can be purified from growth media and concentrated by tangential flow filtration or other filtration methods, and formulated as described above.
  • Intracellular polysaccharides can be also formulated in a similar or identical manner after purification from other cellular components.
  • the invention includes a method of formulating a cosmeceutical composition, said method comprising culturing microalgal cells in suspension under conditions to allow cell division; separating the microalgal cells from culture media, wherein the culture media contains exopolysacchari.de molecules produced by the microalgal cells; separating the exopolysaccharide molecules from other molecules present in the culture media; homogenizing the microalgal cells; and adding the separated exopolysaccharide molecules to the cells before, during, or after homogenization.
  • the microalgal cells are from the genus Porphyridium.
  • polysaccharides both secreted and intracellular, that are suitable for formulation with a carrier for topical application are listed in Table 1.
  • Ratios of homogenatercarrier are typically in the range of about 0.001 : 1 to about 1:1 (volume:volume), although the invention comprises ratios outside of this range, such as, but not limited to, about 0.01:1 and about 0.1:1.
  • Microalgal cellular extracts can also be formulated for topical administration. It is preferable but not necessary that the cells are physically or chemically disrupted as part of the formulation process. For example, cells can be centrifuged from culture, washed with a buffer such as 1.0 mM phosphate buffered saline, pH 7.4, and sonicated. Preferably the cells are sonicated until the cell walls have been substantially disrupted, as can be determined under a microscope. For example, Porphyridium sp. cells can be sonicated using a Misonix sonicator as described in Example 15.
  • Cells can also be dried and ground using means such as mortar and pestle, colloid milling, ball milling, or other physical method of breaking cell walls.
  • cell homogenate can be formulated with carrier and fragrance as described above for polysaccharides.
  • Topical compositions can comprise a portion of a complete composition sold as a single unit. Other portions of the complete compositions can comprise an oral supplement intended for administration as part of a regime for altering skin appearance. Because the top layers of the skin contain dead cells, nutrients delivered via capillaries cannot reach the outer layers of cells. The outer layers of cells must be provided with nutrients though topical administration. However, topical administration is not always an effective method of providing nutrients to deep layers of skin that contain living cells.
  • compositions provided herein comprise both topical compositions that contain algal polysaccharides and/or cellular extracts as well as oral compositions comprising nutraceutical molecules such as purified polysaccharides, whole cell extracts, carotenoids, polyunsaturated fatty acids, and other molecules that are delivered to the skin via capillaries.
  • nutraceutical molecules such as purified polysaccharides, whole cell extracts, carotenoids, polyunsaturated fatty acids, and other molecules that are delivered to the skin via capillaries.
  • topical components of the composition examples include exopolysaccharide from Porphyridium cruentum, Porphyridium sp., list others.
  • Other components of the topical composition can include polysaccharides and/or cell extracts from species listed in Table 1.
  • Cellular extracts for topical administration can also include cellular homogenates from microalgae that have been genetically engineered.
  • homogenates of Porphyridium sp. that have been engineered to express an exogenous gene encoding superoxide dismutase can be formulated for topical administration.
  • Other genes that can be expressed include carotenoid biosynthesis enzymes and polyunsaturated fatty acid biosynthesis enzymes.
  • compositions for oral administration include one or more of the following: DHA, EPA, ARA, lineoileic acid, lutein, lycopene, beta carotene, braunixanthin, zeaxanthin, astaxanthin, linoleic acid, alpha carotene, vitamin C and superoxide dismutase.
  • Compositions for oral administration usually include a carrier such as those described above.
  • Oral compositions can be formulated in tablet or capsule form.
  • Oral compositions can also be formulated in an ingestible form such as a food, tea, liquid, etc.
  • the topical composition and the oral composition both contain at least one molecule in common.
  • the topical composition contains homogenate of Porphyridium cells that contain zeaxanthin, and the oral composition contains zeaxanthin.
  • the topical composition contains homogenate of Porphyridium cells that contain polysaccharide, and the oral composition contains polysaccharide purified from Porphyridium culture media.
  • compositions described herein are packaged for sale as a single unit.
  • a unit for sale comprises a first container holding a composition for topical administration, a second container holding individual doses of a composition for oral administration, and optionally, directions for co-administration of the topical and oral composition.
  • Some embodiments of the invention include a combination product comprising 1) a first topical composition comprising a microalgal extract or other composition and a carrier suitable for topical application to skin; and 2) a second composition comprising at least one compound in common with the first composition and optionally a carrier suitable for human oral consumption; wherein the first and second compositions are packaged for sale as a single unit.
  • the invention includes co-packaging of the two compositions, optionally with a instructions and/or a label indicating the identity of the contents and/or their proper use.
  • the first topical composition is selected from an algae extract, optionally from a species selected from Table 1 , a vitamin C preparation (such as L-ascorbic acid or ascorbyl palmitate), and a vitamin A preparaion (such as d-alpha-tocopherol).
  • a vitamin C preparation such as L-ascorbic acid or ascorbyl palmitate
  • a vitamin A preparaion such as d-alpha-tocopherol
  • the second oral compositions is selcted from an algae extract, optionally from a species selected from Table 1, a vitamin C composition (such as L-ascorbic acid or ascorbyl palmitate), a vitamin A preparaion (such as d-alpha-tocopherol), a polyunsaturated fatty acid (such as Eicosapentaenoic Acid (EPA) or Docosahexaenoic Acid (DHA), zeaxanthm, lutein, beta carotene, and a coenzyme Q preparation (such as ubiquinol-10).
  • a vitamin C composition such as L-ascorbic acid or ascorbyl palmitate
  • a vitamin A preparaion such as d-alpha-tocopherol
  • a polyunsaturated fatty acid such as Eicosapentaenoic Acid (EPA) or Docosahexaenoic Acid (DHA)
  • zeaxanthm such as lutein, beta
  • Prefferred algae extracts contain at least a subset of vitamin C, vitamin A, polyunsaturated fatty acids such as EPA or DHA, and carotenoids such as zeaxanthin, lutein, beta carotene, and cofactors such as coenzyme Q.
  • the invention includes a polysaccharide composition suitable for injection into skin to improve its appearance.
  • the injection is made to alleviate or eliminate wrinkles.
  • the treatment reduces the visible signs of aging and/or wrinkles.
  • human skin As known to the skilled person, human skin, as it ages, gradually loses skin components that keep skin pliant and youthful-looking.
  • the skin components include collagen, elastin, and hyaluronic acid, which have been the subject of interest and use to improve the appearance of aging skin.
  • the invention includes compositions of microalgal polysaccharides, microalgal cell extracts, and microalgal cell homogenates for use in the same manner as collagen and hyaluronic acid.
  • the polysaccharides will be those of from a Porphyridium species, such as one that has been subject to genetic and/or nutritional manipulation to produce polysaccharides with altered monosaccharide content and/or altered sulfation.
  • the polysaccharides are formulated as a fluid, optionally elastic and/or viscous, suitable for injection.
  • the compositions may be used as injectable dermal fillers as one non-limiting example.
  • the injections may be made into skin to fill-out facial lines and wrinkles. In other embodiments, the injections may be used for lip enhancement.
  • the microalgal polysaccharides, cell extracts, and cell homogenates of the invention may be co-formulated with collagen and/or hyaluronic acid (such as the Restylane ® and Hylaform ® products) and injected into facial tissue.
  • hyaluronic acid such as the Restylane ® and Hylaform ® products
  • the polysaccharide is substantially free of protein. The injections may be repeated as deemed appropriate by the skilled practitioner, such as with a periodicity of about three, about four, about six, about nine, or about twelve months.
  • a hyaluronic acid material is mixed with a polysaccharide from the genus Porphyridium prior to co-administration.
  • the invention in this particular embodiment provides longer half-life to the hyaluronic acid due to the potent inhibition of hyaluronidase by polysaccharides isolated from microalgae from the genus Porphyridium. This allows for less injections to a patent.
  • the invention includes a method of cosmetic enhancement comprising injecting a polysaccharide produced by microalgae into mammalian skin.
  • the injection may be of an effective amount to produce a cosmetic improvement, such as decreased wrinkling or decreased appearance of wrinkles as non-limiting examples.
  • the injection may be of an amount which produces relief in combination with a series of additional injections.
  • the polysaccharide is produced by a microalgal species, or two or more species, listed in Table 1.
  • the microalgal species is of the genus Porphyridium and the polysaccharide is substantially free of protein.
  • compositions for non-systemic administration include those formulated for localized administration with little or slow release to other parts of a treated subject's body.
  • Non-limiting examples of non-systemic administration include intravaginal application such as via a suppository, cream or foam; injection into a joint between bones; intravitreous or intraocular administration; and rectal administration via suppository, irrigation or other suitable means.
  • the composition is formulated for the treatment of sexually transmitted diseases, such as those caused by viral agents.
  • Polysaccharides from microalgae provided herein posses potent antiviral activity (see references cited in Table 1).
  • polysaccharides with lubricant properties are used in the practice of certain aspects of the invention. These polysaccharides maybe formulated in solutions that are added to prophylactic devices.
  • the polysaccharides may be one or more described herein, optionally sulfated.
  • the polysaccharide is produced by a microalgal species, or two or more species, listed in Table 1.
  • the microalgae is Porphyridium sp. or Porphyridium omentum.
  • the invention includes a sexually transmitted disease prevention composition, said composition comprising 1) a solution comprising a polysaccharide produced from microalgae; and 2) a prophylactic device.
  • the solution and device are kept separate, but packaged together as a single unit for sale.
  • the solution may be applied to the device by the end user before actual use.
  • the solution and device are packaged so that the solution is in direct contact with the device.
  • the prophylactic devices include, but are not limited to, condoms, sponges, and diaphragms.
  • the devices are packaged with a lubricant.
  • the polysaccharide acts as a lubricant and so no other lubricant is needed.
  • the substance in the composition providing a lubricant function and the substance in the composition providing antiviral activity are the same substance.
  • a combination of a lubricant, such as a cream or lotion, with the polysaccharide of the invention may be used.
  • the polysaccharide is in a composition with a carrier used with a prophylactic device described above.
  • a carrier include a spermicide and a lubricant.
  • a triple composition comprising spermicide, lubricant and the polysaccharide, may be used.
  • the polysaccharide is associated with a fusion (or chimeric) protein comprising a first protein (or polypeptide region) with at least about 60% amino acid identity with the protein of SEQ ID NO: 28.
  • the first protein has at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 98%, or higher, amino acid identity with the sequence of SEQ ID NO:28, and further wherein the fusion protein has the ability to specifically bind to the polysaccharide, preferably with high affinity.
  • the fusion protein may comprise a second protein, or polypeptide region, with a homogenous or heterologous sequence.
  • the second protein is an antibody.
  • the antibody is selective for binding to an antigen of a pathogen, or opportunistic organism, involved in a sexually transmitted disease.
  • Non- limiting examples of antibodies include those that bind an antigen from a pathogen selected from HIV, Herpes Simplex Virus, gonorrhea, Chlamydia, Human Papilloma Virus, and Trichomoniasis .
  • the compositions are formulated for improving joint lubrication or treating joint disorders.
  • microalgal polysaccharides may be used in the same manner as, or in combination with, hyaluronic acid in some compositions of the invention.
  • Hyaluronic acid, or hyaluronan is used to lubricate joints, such as in viscosupplementation.
  • SYNVISC ® Genzyme Corporation
  • SYNVISC ® Genzyme Corporation
  • the elastic and viscous nature of the fluid allows it to function in absorbing shock and improve proper knee movement and flexibility.
  • hyaluronic acid products include Hylartil ® , Arthrease ® , Orthovisc ® and Artval ® .
  • a hyaluronic acid material is mixed with a polysaccharide from the genus Porphyridium prior to injection into a mammalian joint.
  • the invention in this particular embodiment provides longer half-life to the hyaluronic acid due to the potent inhibition of hyaluronidase by polysaccharides isolated from microalgae from the genus Porphyridium. This allows for less injections to a patent.
  • Microalgal polysaccharides of the invention are also formulated as fluids with elastic and/or viscous properties such that they may serve as replacements for normal joint fluid.
  • Polysaccharides from the red microalgae Porphyidium sp. have desirable load bearing and shear properties.
  • the polysaccharides are not degraded by hyaluronidase, which degrades hyaluronic acid; are resistant to elevated temperatures; and are anti-inflammatory and anti-irritating.
  • hyaluronidase which degrades hyaluronic acid
  • the polysaccharides are resistant to elevated temperatures; and are anti-inflammatory and anti-irritating.
  • Golan et al. "Characterization of a Superior Bio-Lubricant Extracted from a Species of Red Microalga " The 39 th Annual Meeting of the Israel Society for Microscopy, Ben Gurion University, May 19 th , 2005, Poster Abstracts (at www.technion.ac.il/technion/materials/ism/ISM2005_posters_abstracts.html); and Gourdon et al.
  • the polysaccharides of the invention may be used in the same or a similar manner, in some embodiments, the polysaccharides will be those from a.
  • Porphyridium species such as one that has been subject to genetic and/or nutritional manipulation to produce polysaccharides with altered monosaccharide content
  • a fluid containing one or more polysaccharides is injected into a joint to.
  • alleviate joint pain such as, but not limited to, arthritis and osteoarthritis.
  • joint pain include pain of the knee, shoulder, elbow, and wrist joints.
  • Subjects afflicted with, suffering from, or having joint pain may be diagnosed and/or identified by a skilled person in the field using any suitable method.
  • Non-limiting examples include signs of inflammation, like swelling, pain, or redness; excess fluid in the joint; the need for physical therapy; pain during exercise.
  • the polysaccharides of the invention are used after the failure, or ineffectiveness, of non- drug treatments or drug therapy for joint pain.
  • non-drug treatments that may be ineffective include avoidance of activities that cause the joint pain, exercise, physical therapy, and removal of excess fluid.
  • drug therapy that may be ineffective include pain relievers, such as acetaminophen and narcotics; anti-inflammatory agents, such as aspirin and other nonsteroidal anti-inflammatory drugs (NSAIDs) such as ibuprofen and naproxen; and injection of steroids.
  • NSAIDs nonsteroidal anti-inflammatory drugs
  • the invention includes a method of mammalian joint lubrication.
  • Mammalian joint lubrication is used to treat conditions such as osteoarthritis, joint trauma, rheumatoid arthritis, and other degenerative conditions affecting the mammalian joint.
  • Mammalian joints include knees, hips, ankles, shoulders, and other joints.
  • the method comprises injecting a microalgal polysaccharide of the invention into a cavity containing synovial fluid.
  • the injection may be of an effective amount to produce relief from one or more symptoms of joint pain or discomfort that is alleviated by joint lubrication. Alternatively, the injection may be of an amount which produces relief in combination with a series of additional injections.
  • the polysaccharide is produced by a microalgal species, or two or more species, listed in Table 1.
  • the microalgal species is of the genus Porphyridium.
  • the methods may also comprise treatment with one or more of the non-drug treatments or drug therapies described herein.
  • injection of a joint lubricating composition of the invention may be combined with administration of an anti-inflammatory agent and optionally physical therapy.
  • polysaccharides can be formulated with carriers, excipients, and other compounds, pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d.alpha-tocopherol polyethyleneglycol 1000 succinate, or other similar polymeric delivery matrices or systems, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxy
  • Cyclodextrins such as alpha-, beta-, and gamma-cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3-hydroxypropyl-beta-cyclodextrins, or other solublized derivatives may also be advantageously used to enhance delivery of therapeutically-effective plant essential oil compounds of the present invention.
  • the polysaccharide compositions of this invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir, however, oral administration or administration by injection is preferred.
  • the pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles.
  • the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • the polysaccharide compositions may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • suitable vehicles and solvents that may be employed are mannitol, water, Ringers solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant such as Ph. HeIv or a similar alcohol.
  • Sterile injectable polysaccharide compositions preferably contain less than 1% protein as a function of dry weight of the composition, more preferably less than 0.1% protein, more preferably less than 0.01% protein, less than 0.001% protein, less than 0.0001% protein, more preferably less than 0.00001% protein, more preferably less than 0.000001% protein.
  • the polysaccharide compositions of the present invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, and aqueous suspensions and solutions.
  • carriers which are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried corn starch.
  • aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • compositions of the present invention may also be administered in the form of suppositories for rectal administration.
  • These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • the invention further provides for the use of polysaccharides as food additives.
  • the polysaccharides are used to aid in keeping food products homogenous and/or thicken food products, such as in a manner analogous to various gums (e.g. xanthan gum or gum Arabic) known to the skilled person.
  • the polysaccharides act as a hydrocolloid polysaccharide and may be used in the same manner as other hydrocolloid polysaccharides. See for example U.S. Patent 5,126,158.
  • the polysaccharides are used to stabilize or emulsify foods.
  • the polysaccharides are based upon their rheological properties, which are similar or superior to those of previously used gums and additives.
  • the polysaccharides will be those of from a Porphyridium species, such as one that has been subject to genetic and/or nutritional manipulation to produce polysaccharides with altered monosaccharide content and/or altered sulfation.
  • the methods of the invention thus include a method comprising addition of one or more polysaccharides of the invention to a food product.
  • the addition may be for use as a thickener or an emulsifier, such as to keep foods homogenous.
  • the addition is to a beverage product, such as to thicken it or improve its texture, appearance, or "feel" upon consumption.
  • the method stabilizes and/or emulsifies a food composition or product.
  • the method may comprise adding one or more microalgal polysaccharide, as described herein, to the food composition or product.
  • Non-limiting examples include polysaccharides produced by microalgae described herein. In some instances, the microalgae is of the genus Porphyridium.
  • a microalgal cell material (such as a homogenate) of the invention may be incorporated into a food, a food supplement, a liquid, a beverage, an emulsion, a powder, or mixtures thereof for ingestion by a mammal.
  • the cell material is formulated as a composition for addition to another food substance, such as for sprinkling, or spreading, on a salad, bread, or cereal as non-limiting examples.
  • the cell material may be incorporated into a cereal product, such as a ready- to-eat variety; incorporated into a baked product, such as a bread, muffin, or waffle; incorporated into a pasta, healthy dessert, snack (such as an athletic bar or healthy drink as non-limiting examples) or a high fiber food.
  • a cereal product such as a ready- to-eat variety
  • a baked product such as a bread, muffin, or waffle
  • incorporated into a pasta, healthy dessert, snack such as an athletic bar or healthy drink as non-limiting examples
  • a high fiber food such as a pasta, healthy dessert, snack (such as an athletic bar or healthy drink as non-limiting examples) or a high fiber food.
  • the invention includes nutraceutical compositions comprising one or more polysaccharides, or microalgal cell extract or homogenate, of the invention.
  • a nutraceutical composition serves as a nutritional supplement upon consumption.
  • a nutraceutical may be bioactive and serve to affect, alter, or regulate a bioactivity of an organism.
  • a nutraceutical may be in the form of a solid or liquid formulation.
  • a solid formulation includes a capsule or tablet formulation as described above.
  • a solid nutraceutical may simply be a dried microalgal extract or homogenate, as well as dried polysaccharides per se.
  • the invention includes suspensions, as well as aqueous solutions, of polysaccharides, extracts, or homogenates.
  • the methods of the invention include a method of producing a nutraceutical composition.
  • a method may comprise drying a microalgal cell homogenate or cell extract.
  • the homogenate may be produced by disruption of microalgae which has been separated from culture media used to propagate (or culture) the microalgae
  • a method of the invention comprises culturing red microalgae; separating the microalgae from culture media; disrupting the microalgae to produce a homogenate; and drying the homogenate.
  • a method of the invention may comprise drying one or more polysaccharides produced by the microalgae.
  • a method of the invention comprises drying by tray drying, spin drying, rotary drying, spin flash drying, or lyophilization.
  • methods of the invention comprise disruption of microalgae by a method selected from pressure disruption, sonication, and ball milling
  • a method of the invention further comprises formulation of the homogenate, extract, or polysaccharides with a carrier suitable for human consumption.
  • the formulation may be that of tableting or encapsulation of the homogenate or extract.
  • the methods comprise the use of microalgal homogenates, extracts, or polysaccharides wherein the cells contain an exogenous nucleic acid sequence, such as in the case of modified cells described herein.
  • the exogenous sequence may encode a gene product capable of being expressed in the cells or be a sequence which increases expression of one or more endogenous microalgal gene product.
  • Non-limiting examples of the latter include insertion of regulator regions which increase expression of an endogenous microalgal gene and insertion of additional copies of an endogenous microalgal gene to increase copy number.
  • some embodiments of the invention include microalgal cells expressing an exogenous gene which increases production of a small molecule naturally produced by the microalgae or which induces the microalgae to produce, or directs the production of, a small molecule not naturally produced by the microalgae.
  • the increased expression of an endogenous microalgal gene or insertion of additional copies of an endogenous microalgal gene to increase copy number is used to increase production of a small molecule normally produced by the microalgae.
  • the microalgal homogenates, extracts, or polysaccharides are from cells containing a modification to an endogenous nucleic acid sequence.
  • One non-limiting example includes modified microalgal cells wherein an endogenous repressor nucleic acid sequence, or sequence encoding a proteinaceous or RNA gene product, is removed or inhibited such that production of a small molecule normally produced by the microalgae is increased.
  • nucleic acid modification as described herein increases production of more than one microalgal small molecule.
  • the small molecule of a microalgal cell which is increased by these methods of the invention is a carotenoid.
  • carotenoids include lycopene, lutein, beta carotene, zeaxanthin.
  • the small molecule is a polyunsaturated fatty acid, such as, but not limited to, EPA, DHA, liholeic acid and ARA.
  • the invention includes a nutraceutical composition prepared by a method described herein, hi some embodiments, the composition comprises homogenized red microalgal cells and a carrier suitable for human consumption, hi other embodiments, the carrier is a food product or composition.
  • the microalgal cells may be genetically modified as described above to result in red microalgae which produce an increased amount of a small molecule naturally produced by the red microalgae; or to produce a small molecule not naturally produced by the microalgae.
  • the small molecule is DHA.
  • the invention further provides for a combination composition wherein a microalgal homogenate further comprises an exopolysaccharide produced by the red microalgae.
  • the exopolysaccharide has been purified from culture media used to grow the red microalgae.
  • the exopolysaccharide may be added to the cells before, during, or after homogenization.
  • a microalgal homogenate further comprises an exogenously added molecule, such as, but not limited to, EPA, DHA, linoleic acid, ARA 5 lycopene, lutein, beta carotene, and zeaxanthin.
  • a nutraceutical of the invention may also be a composition comprising a purified first polysaccharide produced from a microalgal species listed in Table 1 and a carrier suitable for human consumption.
  • the polysaccharides include sulfated molecules as well as polysaccharides with an average molecular weight (MW) of the polysaccharide is between about 2 and about 7 million Daltons (MDa).
  • MW average molecular weight
  • the polysaccharide has an average MW of about 3, about 4.5, about 5, or about 6 MDa.
  • the average MW is below 2 MDa 5 such as below about 1, below about 0.8, below about 0.6, below about 0.4, or below about 0.2 MDa.
  • the composition contains between 1 microgram and 50 grams of one type of microalgal polysaccharide.
  • the composition contains more than one type of microalgal polysaccharide, such as one or more additional polysaccharide.
  • at least one polysaccharide is optionally from a non-microalgal source, such as a non-microalgal species.
  • the additional polysaccharide is beta glucan.
  • a composition further comprises a plant phytosterol.
  • a composition comprising both a microalgal homogenate and a polysaccharide, such as an exopolysaccharide, is disclosed herein.
  • the composition may comprise homogenized microalgae and isolated or purified or semi-purified exopolysaccharide(s), wherein the composition is a percentage of exopolysaccharide by weight ranging from up to about 1% to up to about 20%, or higher.
  • the remaining portion of the composition may be the homogenate or other carriers and excipients as desired for a composition, nutraceutical, or cosmeceutical of the invention.
  • the percentage of exopolysaccharide is up to about 2%, up to about 5%, or up to about 10%.
  • This type of combination composition may be prepared by any appropriate means known to the skilled person, including preparing of each component separately and then combining them.
  • formulation of a composition comprises subjected a microalgal culture containing exopolysaccharides to tangential flow filtration to concentrate the material and then diafiltration until the composition is substantially free of salts, wherein the cells and exopolysaccharide are both retained in the retentate.
  • the material can also be partially concentrated, diafiltered, and then concentrated further, and this regime can also be used on supernatant free of cells where the exopolysaccharide is retained.
  • the exopolysaccharides may be those produced by the microalgae during culture or may be exogenously added to the culture before processing.
  • the filtered material may then be homogenized or dried as described herein.
  • a combination of a first composition for topical application and second composition for consumption is provided.
  • the first composition may be a topical formulation or non-systemic formulation, optionally a cosmeceutical, as described herein.
  • the first composition comprises a carrier suitable for topical application to skin, such as human skin.
  • the second composition include a food composition or nutraceutical as described herein.
  • the second composition comprises at least one carrier suitable for human consumption, such as that present in a food product or composition.
  • the first and second compositions contain at least one compound in common.
  • Non-limiting examples include one polysaccharide or one carrier in common.
  • the at least one compound is selected from DHA, EPA, ARA, lycopene, lutein, beta carotene, zeaxanthin, linoleic acid, vitamin C, and superoxide dismutase.
  • Combination products of the invention may be packaged separately for subsequent use together by a user or packaged together to facilitate purchase and use by a consumer.
  • Packaging of the first and second compositions may be for sale as a single unit.
  • a polysaccharide (as well as homogenate or extract) containing food product or nutraceutical of the invention maybe consumed as a source of nutrition and/or sustenance.
  • the invention includes methods of providing food, nutrition or sustenance to a subject, such as a human being, by administration of a composition or nutraceutical as described herein. While a food product may be a primary source of sustenance, a nutraceutical may be used as a nutritional supplement.
  • the invention also includes methods of administering both to a subject.
  • the administered food product may comprise a polysaccharide, extract, or homogenate as described herein.
  • the polysaccharide, extract or homogenate is used to thicken, stabilize or emulsify foods.
  • a polysaccharide containing composition including those containing a microalgal homogenate or extract of the invention.
  • the composition is used to regulate, or aid in the regulation of insulin.
  • Administration of algal polysaccharides included in the invention reduces insulin secretion in response to a given stimulus.
  • Subjects, including human beings, in need of insulin regulation may be identified by any means known to the skilled person.
  • the subject is identified as being at risk for diabetes by a skilled clinician. Being at risk includes having one or more risk factors, as assessed by the skilled . person, which increase the chances of needing insulin regulation and/or having diabetes.
  • risk factors include those of lifestyle, behavior, health status, disease, and medication use.
  • the risk factors may amount to the present of "pre-diabetes" or "metabolic disease".
  • Non-limiting examples of lifestyle factors include inactivity, stress, diet, and aging.
  • Non-limiting examples of behavior factors include levels of sexual activity, smoking, alcohol use, and drug use.
  • Non-limiting examples of health status factors include obesity, cholesterol, diabetes, immunosuppression, and hypertension as well as gender status as a woman, such as pregnancy, childbirth, and menopause.
  • the compositions are particularly useful for lowering cholesterol levels in patients having abnormally high levels of cholesterol of at least 240 mg/dL total cholesterol, at least 160 mg/dL LDL cholesterol, no more than 40 mg/dL HDL cholesterol, and/or at least 400 mg/dL triglycerides.
  • Non-limiting examples of diseases include HIV, heart, cancer, and autoimmune diseases.
  • Non-limiting examples of medications include use of contraceptives and steroids.
  • a nutraceutical of the invention may be administered to a subject found to have one or more of these risk factors sufficient to warrant conservative or aggressive treatment of the subject.
  • the determination or diagnosis of risk factor presence may be conducted by a skilled person, such as a clinician.
  • Non-limiting examples of conservative treatment methods may comprise administration of a polysaccharide composition of the invention optionally in combination of one or more alterations in activity to reduce one or more risk factors.
  • the methods may be in the absence of other treatment for insulin malfunction or misregulation, pre-diabetes, or metabolic disease.
  • Non-limiting examples of aggressive treatment include active administration of a bioactive agent to a subject afflicted with diabetes or insulin misregulation or malfunction.
  • Administration of a bioactive agent includes insulin injection to maintain glucose levels in a subject.
  • a method of regulating insulin may comprise administering a polysaccharide produced by microalgae as described herein.
  • the polysaccharides may reduce the need for other agents, such as a bioactive agent, that regulate insulin.
  • antioxidant properties of microalgal polysaccharides may be utilized to treat subjects in need of antioxidant activity.
  • Polysaccharides with antioxidant activity may be identified by.suitable means known to the skilled person.
  • the polysaccharides will be those from a Porphyridium species, such as one that has been subject to genetic and/or nutritional manipulation to produce polysaccharides with altered monosaccharide content and/or altered sulfation.
  • antioxidant polysaccharides are used to inhibit, reduce or treat undesired inflammation.
  • the inflammation can be the result of several diseases including autoimmune diseases, graft versus host disease, host versus graft disease, or pathogenic infections.
  • the polysaccharides will be those from a Porphyridium species, such as one that has been subject to genetic and/or nutritional manipulation to produce polysaccharides with altered monosaccharide content and/or altered sulfation.
  • aspects of the invention include a method of reducing reactive oxygen species (ROS) in a mammal.
  • the method is used to prevent or treat a disease or unwanted condition associated with ROS or oxidative stress.
  • a disease or unwanted condition include a neurodegenerative condition, atherosclerosis, metabolic syndrome, erectile dysfunction, transplant rejection, cancer, cardiovascular disease, hypertension, ischemia, reperfusion injury, rheumatoid arthritis, and aging, which are all conditions that are associated with increased in reactive oxygen species.
  • a method comprises administering a composition comprising red microalgae cell material (such as a homogenate) as described herein.
  • the composition comprises one or more other agents or compounds with antioxidant activity.
  • the method may be used to lower the level of ROS, or reduce or decrease the amount of damage caused by ROS.
  • the amount of the composition may be any that is effective or sufficient to produce a desired improvement or therapeutic benefit.
  • the method may be used to prevent, treat, or alleviate a symptom of, a neurodegenerative disease or disorder.
  • Non-limiting examples of a neurodegenerative disease or disorder include dementias (e.g., senile dementia, memory disturbances/memory loss, dementias caused by neurodegenerative disorders (e.g., Alzheimer's, Parkinson's disease, Parkinson's disorders, Huntington's disease (Huntington's Chorea), Freidreich's ataxia, Lou Gehrig's disease, multiple sclerosis, Pick's disease, Parkinsonism dementia syndrome), progressive subcortical gliosis, progressive supranuclear palsy, thalmic degeneration syndrome, hereditary aphasia, amyotrophic lateral sclerosis, Shy-Drager syndrome, and Lewy body disease; vascular conditions (e.g., infarcts, hemorrhage, cardiac disorders); mixed vascular and Alzheimer's; bacterial meningitis; Creutzfeld-Jacob Disease; and Cushing's disease.
  • the method may be used to delay the onset of one or more of the above diseases or
  • the method may also be used to prevent, treat, or alleviate metabolic syndrome in a human subject, which is characterized by at least three of the following: enlargement of the waist diameter, a higher level of arterial pressure, a higher level of low density lipoprotein cholesterol, a higher level of glycemia, and reduction of high density lipoprotein cholesterol (see for example, Palomo et al. Int. J. MoL Med. (2006) 18(5):969-74).
  • the method may also be used to prevent, treat, or alleviate a symptom of, erectile dysfunction.
  • the involvement of ROS in the disorder has been reported by Jeremy et al. (Int. J. Impot. Res. 2006 Oct 19; Epub ahead of print).
  • the method may also be used to prevent, treat, or alleviate a symptom of, transplant rejection, such as in the case of organ or tissue transplant as non-limiting examples.
  • transplant rejection such as in the case of organ or tissue transplant as non-limiting examples.
  • the involvement of ROS in transplant rejection is known to the skilled person. See for example Cell Immunol. 2006 Jun;241(2): 59-65.
  • the method may also be used to prevent, treat, or alleviate a symptom of aging.
  • the involvement of ROS in aging has been reported by Valko et al. (Int. J. Biochem Cell Biol. (2007) 39(l):44-84 Epub 2006 Aug 4).
  • the invention includes a method to treat inflammation.
  • a method may comprise administering a polysaccharide containing composition of the invention to a subject in need of anti-inflammatory activity.
  • the polysaccharide may be one or more produced by microalgae described herein.
  • the administering may be by a variety of means, including direct transfer to a tissue or subject via an intramuscular, intradermal, subdermal, subcutaneous, oral, parenteral, intraperitoneal, intrathecal, or intravenous procedure.
  • a scaffold or binding protein can be placed within a cavity of the body, such as during surgery, or by inhalation, or vaginal or rectal administration.
  • compositions or medicaments are administered to a patient susceptible to, or otherwise at risk of, a disease or condition, such as excess cholesterol, inflammation, low insulin, inadequate joint lubrication in an amount sufficient to eliminate or reduce the risk, lessen the severity, or delay the outset of the disease, including biochemical, histologic and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
  • a disease or condition such as excess cholesterol, inflammation, low insulin, inadequate joint lubrication in an amount sufficient to eliminate or reduce the risk, lessen the severity, or delay the outset of the disease, including biochemical, histologic and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
  • compositions or medicants are administered to a patient suspected of, or already suffering from such a disease in an amount sufficient to cure, or at least partially arrest, the symptoms of the disease (biochemical, histologic and/or behavioral), including its complications and intermediate pathological phenotypes in development of
  • a microalgal cell material containing composition of the invention may also be used in an effective or sufficient amount to produce a desired improvement or therapeutic benefit in decreasing mammalian body fat (such as via a hypolipidemic effect), controlling or preventing weight gain in a mammal, reducing the body weight in a mammal, increasing satiety (or decreasing appetite) in a mammal, increasing energy expenditure (or energy use) in a mammal, reducing inflammation in a mammal, or preventing and/or treating atherosclerosis in a mammal.
  • mammalian body fat such as via a hypolipidemic effect
  • compositions comprising red microalgae cell material (such as a homogenate) for ingestion by a mammal as described herein.
  • the composition comprises one or more other agents or compounds with anti-oxidant activity.
  • Genes can be expressed in microalgae by providing, for example, coding sequences in operable linkage with promoters.
  • An exemplary vector design for expression of a gene in microalgae contains a first gene in operable linkage with a promoter active in algae, the first gene encoding a protein that imparts resistance to an antibiotic or herbicide.
  • the first gene is followed by a 3' untranslated sequence containing a polyadenylation signal.
  • the vector may also contain a second promoter active in algae in operable linkage with a second gene.
  • the second gene can encode any protein, for example an enzyme that produces small molecules or a mammalian growth hormone that can be advantageously present in a nutraceutical.
  • codon-optimized cDNAs for methods of recoding genes for expression in microalgae, see for example US patent application 20040209256.
  • promoters in expression vectors are active in algae, including both promoters that are endogenous to the algae being transformed algae as well as promoters that are not endogenous to the algae being transformed (ie: promoters from other algae, promoters from plants, and promoters from plant viruses or algae viruses).
  • Example of methods for transforming microalgae in addition to those demonstrated in the Examples section below, including methods comprising the use of exogenous and/or endogenous promoters that are active in microalgae, and antibiotic resistance genes functional in microalgae, have been described. See for example; Curr Microbiol. 1997 Dec;35(6):356-62 (Chlorella vulgaris); Mar Biotechnol (NY).
  • SEQ ID NO:44 a promoter that drives the gene encoding the glycoprotein that binds the polysaccharide from Porphyridium sp. (see GenBank accession number AY778963 for the cDNA of this gene).
  • the promoter of SEQ ID NO:44 is a preferred promoter for driving exogenous genes in algae of the genus Porphyridium.
  • Suitable promoters may be used to express a nucleic acid sequence in microalgae.
  • the sequence is that of an exogenous gene or nucleic acid.
  • the exogenous gene is one that encodes a carbohydrate transporter protein.
  • Such a gene may be advantageously expressed in a microalgal cell to allow entry of a monosaccharide transported by the transporter protein.
  • the exogenous gene can encode a superoxide dismutase or a mammalian growth hormone.
  • the codon usage may be optionally optimized in whole or in part to facilitate expression in microalgae.
  • the invention includes cells of the genus Porphyridium that have been stably transformed with a vector containing a selectable marker gene in operable linkage with a promoter active in microalgae. In other embodiments the invention includes cells of the genus Porphyridium that have been stably transformed with a vector containing a- selectable marker gene in operable linkage with a promoter endogenous to a member of the Rhodophyte phylum.
  • promoters include SEQ ID NOs: 44 and 55, promoters from the genome of Chondrus crispus (Genbank accession number Z47547), promoters from the genome of Cyanidioschyzon merolae (see for example Matsuzaki, M. et al.
  • the invention includes cells of the genus Porphyridium that have been stably transformed with a vector containing a selectable marker gene in operable linkage with a promoter other than a CMV promoter such as that found in PCT application WO2006013572.
  • the invention thus includes, in some embodiments, a microalgal cell comprising an exogenous gene that encodes a carbohydrate transporter protein.
  • the cell may be that of the genus Porphyridum as a non-limiting example.
  • Non-limiting examples of genes encoding carbohydrate transporters to facilitate the uptake of exogenously provided carbohydrates include genes encoding the proteins of SEQ ID NOs: 20, 22, 24, 26, 27, 29-39 and 46-48 as provided herein.
  • the nucleic acid sequence encodes a protein with at least 60% amino acid sequence identity with a protein with a sequence represented by one of SEQ ID NOs: 20, 22, 24, 26, 27, and 29-39 and 46-48 wherein the protein is located in the plasma membrane of the cell and transports a carbohydrate from the culture media into the cell.
  • the nucleic acid sequence encodes a protein with at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 98%, or higher, amino acid identity with a sequence of these SEQ ID NOs: 20, 22, 24, 26 and 27.
  • the nucleic acid sequence has at least 60% nucleotide identity with a nucleic acid molecule with a one of SEQ ED NOs: 21, 23 and 25.
  • the nucleic acid sequence has at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 98%, or higher, nucleic acid identity with a sequence of these SEQ ID NOs.
  • Hexokinases can also be expressed to facilitate utilization of exogenously provided carbohydrates through phosphorylation of monosaccharides.
  • a nonlimiting example of a hexokinase is SEQ IS NO: 71 or a protein with at least 60% amino acid identity with SEQ ID NO:71 that phosphorylates a monosaccharide.
  • the invention provides for the expression of a protein sequence found to be tightly associated with microalgal polysaccharides.
  • a protein sequence found to be tightly associated with microalgal polysaccharides.
  • One non-limiting example is the protein of SEQ ID NO: 28, which has been shown to be tightly associated with, but not covalently bound to, the polysaccharide from Porphyridium sp. (see J. Phycol. 40: 568-580 (2004)).
  • Porphyridium culture media is subjected to tangential flow filtration using a filter containing a pore size well in excess of the molecular weight of the protein of SEQ ID NO: 28, the polysaccharide in the retentate contains detectable amounts of the protein, indicating its tight association with the polysaccharide.
  • the calculated molecular weight of the protein is approximately 58kD, however with glycosylation the protein is approximately 66 kD.
  • Such a protein may be expressed directly such that it will be present with the polysaccharides of the invention or expressed as part of a fusion or chimeric protein as described herein.
  • a fusion protein the portion that is tightly associated with a microalgal polysaccharide effectively links the other portion(s) to the polysaccharide.
  • a fusion protein may comprise a second protein or polypeptide, with a homogenous or heterologous sequence.
  • a homogenous sequence would result in a dimer or multimer of the protein while a heterologous sequence can introduce a new functionality, including that of a bioactive protein or polypeptide.
  • Non-limiting examples of the second protein include an antibody, a growth hormone or factor, and an enzyme.
  • the enzyme is superoxide dismutase, such as that has at least about 60% amino acid identity with the sequence of SEQ ID NO: 14 or SEQ ID NO: 15 as non-limiting examples.
  • Superoxide dismutase scavenges reactive oxygen species such as the superoxide anion.
  • the superoxide dismutase has at least about 70%, at least about 75%, at least about 80%,. at least about 85%, at least about 90%, at least about 95%, or at least about 98%, or higher, amino acid identity with the sequence of SEQ ID NO: 14 or 15 or a sequence from Table 16.
  • the enzyme is a phytase (such as GenBank accession number CAB91845 and US Patents 6,855,365 and 6,110,719).
  • a fusion between the polysaccharide binding protein and antibodies that specifically bind to and neutralize a pathogen are included in the invention.
  • Non-limiting examples include anti-HIV antibodies, like the 2Gl 2 antibody (see Proc Natl Acad Sci U S A. 2005 Sep 20;102(38): 13372-7); the 1RHH_B antibody (see Clin Exp Immunol. 2005 Jul;141(l):72-80); the scFvlO2 antibody (see J Gen Virol. 2005 Jun;86(Pt 6):1791-800); and the micro Ab antibody (see Nat Med. 2005 Jun;l l(6):615-22; 2Gl 2, 2F5, 4E10, 2gl2 Fab 1ZLS_L).
  • antibodies preferably antibodies that specifically bind to infectious disease agents, can also be expressed in algae without being fused to any other proteins.
  • the biomass containing the recombinant antibodies can be administered orally to deliver the antibodies to a mammal for prophylaxis or treatment.
  • One advantage to a fusion is that the bioactivity of the polysaccharide and the bioactivity from the protein can be combined in a product without increasing the manufacturing cost over only purifying the polysaccharide.
  • the potent antioxidant properties of a Porphyridium polysaccharide can be combined with the potent antioxidant properties of superoxide dismutase in a fusion, however the polysaccharide:superoxide dismutase combination can be isolated to a high level of purity using tangential flow filtration.
  • the potent antiviral properties of s can be combined with the potent antioxidant properties of superoxide dismutase in a fusion, however the polysaccharide:superoxide dismutase combination can be isolated to a high level of purity using tangential flow filtration.
  • Porphyridium polysaccharide can be added to the potent neutralizing activity of recombinant antibodies fused to the protein (SEQ ID NO:28) that tightly associates with the polysaccharide.
  • SEQ ID NO:28 protein that tightly associates with the polysaccharide.
  • fusion proteins of SEQ ID NO:28 or sequences with at least 60% amino acid identity with SEQ ID NO:28 bind with high affinity to a sulfated exopoly saccharide from a cell of the genus Porphyridium. It is preferred but not becessary that the binding be selective.
  • the exogenous gene can encode a protein that catalyzes the conversion of one carotenoids/xanthophyll to another, such as a beta-carotene hydroxylase (beta carotene to zeaxanthin), a lycopene cyclase (lycopene to beta carotene), and other enzymes known in the art.
  • a beta-carotene hydroxylase beta carotene to zeaxanthin
  • lycopene cyclase lycopene to beta carotene
  • the codon usage may be optionally optimized in whole or in part to facilitate expression in microalgae.
  • Beta carotene hydroxylases exhibit a wide degree of amino acid identity despite the fact that that are capable of catalyzing the same reaction.
  • the invention includes cloning and expression of enzymes that catalyze the conversion of beta carotene to zeaxanthin that have at least 25% amino acid identity to those in Table 4.
  • the beta carotene hydroxylases of the invention have at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, and at least 99% amino acid identity to those in Table 4.
  • the invention includes cloning and expression of enzymes that catalyze the conversion of lycopene to beta carotene (ie: lycopene cylases).
  • the lycopene cylases of the invention have at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, and at least 99% amino acid identity to GenBank accession numbers AAA81880, NP_624526, BAA20275, and P21687.
  • Beta carotene hydroxylases exhibit widely divergent amino acid identity while exhibiting the same catalytic activity.
  • BLAST comparisons of beta carotene hydroxylases show as little as 25% amino acid identity: NP_682690 vs. NP_896386 (59% identity); ABB49299 vs. YP_172377 (67% identity); NP_682690 vs. AAT38625 (25% identity); YP_457405 vs. NP_440788 (25% identity); ZPJ)1084736 vs. NP_848964 (55% identity).
  • BLAST parameters used in the analysis were default parameters (blastp program, matrix BLOSUM62, open gap 11 and extension gap I 9 gapx_dropoff 50, expect 10.0, word size 3, filter activated).
  • the invention includes genetic expression methods comprising the use of an expression vector.
  • a microalgal cell such as a Porphyridium cell
  • the expression vector may comprise a resistance cassette comprising a gene encoding a protein that confers resistance to an antibiotic, such as zeocin, or another selectable marker such as a carbohydrate transporter gene for selection in the dark in the presence of a fixed carbon source, operably linked to a promoter active in microalgae.
  • the vector may also comprise a second expression cassette comprising a second protein to a promoter active in microalgae. The two cassettes are physically linked in the vector.
  • the transformed cells may be optionally selected based upon the ability to grow in the presence of the antibiotic or other selectable marker under conditions wherein cells lacking the resistance cassette would not grow, such as in the dark.
  • the resistance cassette, as well as the expression cassette may be taken in whole or in part from another vector molecule.
  • a method of expressing an exogenous gene in a cell of the genus Porphyridium may comprise operably linking a gene encoding a protein that confers resistance to the antibiotic zeocin to a promoter active in microalgae to form a resistance cassette; operably linking a gene encoding a second protein to a promoter active in microalgae to form a second expression cassette, wherein the resistance cassette and second expression cassette are physically connected to form a dual expression vector; transforming the cell with the dual expression vector; and selecting for the ability to survive in the presence of at least 2.5 ug/ml zeocin, preferably at least 3.0 ug/ml zeocin., and more preferably at least 3.5 ug/ml zeocin, more preferably at least 5.0 ug/ml zeocin.
  • the expression cassette expresses a growth hormone, optionally mammalian and optionally a fish or other vertebrate growth hormone.
  • a growth hormone optionally mammalian and optionally a fish or other vertebrate growth hormone.
  • the growth hormone include bovine growth hormone, human growth hormone, porcine growth hormone, and equine growth hormone.
  • growth hormones include proteins with sequences represented as SEQ ID NOs: 17, 18 and 19.
  • the growth hormone has at least about 60%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 98%, or higher, amino acid identity with a protein sequence represented by one of SEQ ID NOs: 17, 18, 19 or one of Genbank accession numbers AAW67479; AAW67480; AAB641 17; BAC07248; AAF61751; AAX77220; ABA55647; ABI97975; BAA06379; AAU93606; AAX31661; NP_001003168, further wherein the protein stimulates growth in a vertebrate organism.
  • the expression cassette expresses a phytase enzyme, such as for example, SEQ ID NOs: 40 and 41.
  • the invention provides the cells, such as Porphyridium cells, prepared by the above methods.
  • the cells may comprise a mammalian growth hormone via recombinant protein expression as provided above.
  • transgenic Porphyridiwn expressing a mammalian growth hormone and /or a phytase enzyme are formulated into livestock food and administered to animals.
  • the cells may be advantageously used as, or as a component of, animal feed.
  • the advantage to expressing such growth hormones in microalgae such as Porphyridium is that the polysaccharide is not hydro lyzed by the stomach of the mammal (see for example Br J Nutr. 2000 Oct;84(4):469-76), and therefore the cell wall, which is made primarily of polysaccharide, protects the mammalian growth hormone as the cells transit through the stomach and into the intestines. Once in the intestines, the cell wall eventually begins breaking down, allowing the growth hormones to cross into the bloodstream and achieve a pharmacological effect.
  • genes that can be expressed using the methods provided herein encode enzymes that produce nutraceutical small molecules such as lutein, zeaxanthin, and DHA.
  • the genes encoding the proteins are synthetic and are created using preferred codons on the microalgae in which the gene is to be expressed.
  • enzyme capable of turning EPA into DHA are cloned into the microalgae Porphyridium sp. by recoding genes to adapt to Porphyridium sp. preferred codons.
  • enzymes in the carotenoid pathway see SEQ ID NOs: 12 and 13 and sequences from Table 15.
  • sequence comparison For sequence comparison to determine percent nucleotide or amino acid identity, typically one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are input into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated.
  • sequence comparison algorithm calculates the percent sequence identity for the test sequence(s) relative to the reference sequence, based on the designated program parameters.
  • Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith & Waterman, Adv. Appl. Math. 2:482 (1981), by the homology alignment algorithm of Needleman & Wunsch, J. MoI. Biol.
  • HSPs high scoring sequence pairs
  • initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them.
  • the word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always > 0) and N (penalty score for mismatching residues; always ⁇ 0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the default parameters of the BLAST programs are suitable.
  • the BLASTP program uses as defaults a word length (W) of 3, an expectation (E) of 10, and the BLOSUM62 scoring matrix.
  • the TBLATN program (using protein sequence for nucleotide sequence) uses as defaults a word length (W) of 3., an expectation (E) of 10, and a BLOSUM 62 scoring matrix, (see Henikoff & Henikoff, Proc. Natl. Acad. ScL USA 89:10915 (1989)).
  • the BLAST algorithm In addition to calculating percent sequence identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin & Altschul, Proc. Nat'l. Acad. ScL USA 90:5873-5787 (1993)).
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • P(N) the smallest sum probability
  • a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001.
  • microalgae generally have the ability to live off a fixed carbon sources such as glucose, but many do not have transporters that allow for uptake of the fixed carbon source from the culture media.
  • Microalgae cells can be transformed with a gene that encodes a plasma membrane sugar transporter that allows for the selection of growth in the dark, in the absence of photosynthesis, in the presence of the transporter's substrate sugar.
  • Such transformed cells provide a significant benefit in that the need for light energy is reduced or eliminated because the cells may grow and produce cellular products, including polysaccharides, in the presence of fixed carbon material as the energy source. See for example, Science. 2001 Jun 15;292(5524):2073-5. Such growth achieves much higher cell densities in a shorter period of time than photoautotrophic growth.
  • the transformed microalgal cell may be one that is described above as expressing a sugar transporter.
  • Nucleic acids and vectors for such expression are also described above.
  • nucleic acids encoding carbohydrate transporters such as SEQ ID NOs: 20, 22, 24, 26, 27, 29-39 and 46-48 are placed in operable linkage with a promoter active in microalgae.
  • the nucleic acid encoding a carbohydrate transporter contains preferred codons of the organism the vector is transformed into.
  • the nucleic acids of SEQ ID NOs: 21, 23, and 25 encode the carbohydrate transporter proteins of SEQ ID NOs: 20, 22, and 24, respectively.
  • a codon-optimized cDNA encoding a carbohydrate transporter protein, optimized for expression in Porphyridium sp. is placed in operable linkage with a promoter and 3'UTR active in microalgae.
  • the vector is used to transform a cell of the genus Porphyridium using methods disclosed herein, including biolistic transformation, electroporation, and glass bead transformation.
  • a preferred promoter is active in more than one species of microalgae, such as for example the Chlamydomonas reinhardtii RBCS2 promoter (SEQ ID NO:43).
  • any promoter active in microalgae can be used to express a gene in such constructs, and preferred promoters such as RBCS2 and viral promoters have been shown to be active in multiple species of microalgae (see for example Plant Cell Rep. 2005 Mar;23(10-l l):727-35; J Microbiol. 2005 Aug;43(4):3 ⁇ l-5; Mar Biotechnol (NY). 2002 Jan;4(l):63-73).
  • Promoters, cDNAs, and 3'UTRs, as well as other elements of the vectors can be generated through cloning techniques using fragments isolated from native sources (see for example Molecular Cloning: A Laboratory Manual, Sambrook et al. (3d edition, 2001, Cold Spring Harbor Press; and U.S. Patent 4,683,202). Alternatively, elements can be generated synthetically using known methods (see for example Gene. 1995 Oct 16;164(l):49-53),
  • cells may be mutagenized and then selected for the ability to grow in the absence of light energy but in the presence of a fixed carbon source.
  • the invention includes a method of producing microalgal cells that have gained the ability to grow via a fixed carbon source in the absence of photosynthesis. This may also be referred to as trophic conversion of a microalgal cell to no longer be an obligate photoautotroph.
  • the method comprises identifying or selecting cells that have gained the ability to utilize energy from a fixed carbon source.
  • the methods comprise selecting microalgal cells, such as a Porphyridium cell, for the ability to undergo cell division in the absence of light, or light energy.
  • the cells such as one from a species listed in Table 1 , may be those which have been transformed with a sugar transporter or those which have been mutagenized, chemically or non-chemically. The selection may be, for example, on about 0.1% or about 1% glucose, or another fixed carbon source, in the dark. Preferred fixed carbon compounds are listed in Tables 2 and 3.
  • Non-limiting examples of carbohydrate transporter proteins optionally operably linked to promoters active in microalgae, as well as expression cassettes and vectors comprising them, have been described above.
  • the nucleic acids may be incorporated into the genome of a microalgal cell such that an endogenous promoter is used to express the transporter.
  • Additional embodiments of the methods include expression of transporters of a carbohydrate selected from Table 2 or 3.
  • Non-limiting examples of mutagenesis include contact or propagation in the presence of a mutagen, such as ultraviolet light, nitrosoguanidine, and/or ethane methyl sulfonate (EMS).
  • a mutagen such as ultraviolet light, nitrosoguanidine, and/or ethane methyl sulfonate (EMS).
  • a method of the invention comprises providing a nucleic acid encoding a carbohydrate transporter protein; transforming a Porphyridium cell with the nucleic acid; and selecting for the ability to undergo cell division in the absence of light or in the presence of a carbohydrate that is transported by the carbohydrate transporter protein.
  • a method comprises subjecting a microalgal cell to a mutagen; placing the cell in the presence of a molecule listed in Tables 2 or 3; and selecting for the ability to undergo cell division in the absence of light.
  • the methods may also be considered to be for trophically converting a microalgal cell to no longer be an obligate phototroph. It is pointed out that the ability to select for loss of obligate phototrophism also provides an alternative means to select for expression of a sugar transporter in the absence of a selectable marker because correct expression and functionality of the transporter is the selectable phenotype when cells are grown in the absence of light for photosynthesis.
  • Porphyridium sp. (strain UTEX 637) and Porphyridium cruentum (strain UTEX 161) were inoculated into autoclaved 2 liter Erlenmeyer flasks containing an artificial seawater media: 1495 ASW medium recipe from the American Type Culture Collection, (components are per 1 liter of media) NaCl: 27.Og; MgSO 4 • 7H 2 O: 6.6g; MgCl 2 . 6H 2 O: 5.6g; CaCl 2 .
  • Porphyridium sp. and Porphyridium omentum cultures were centrifuged at 4000 rcf. The supernatant was subjected to tangential flow filtration in a Millipore Pellicon 2 device through a 100OkD regenerated cellulose membrane (filter catalog number P2C01MC01). Approximately 4.1 liters of Porphyridium cruentum and 15 liters of Porphyridium sp. supernatants were concentrated to a volume of approximately 200 ml in separate experiments. The concentrated exopolysaccharide solutions were then diafiltered with 10 liters of ImM Tris (pH 7.5).
  • the retentate was then flushed with ImM Tris (pH 7.5), and the total recovered polysaccharide was lyophilized to completion. Yield calculations were performed by the dimethylmethylene blue (DMMB) assay.
  • DMMB dimethylmethylene blue
  • the lyophilized polysaccharide was resuspended in deionized water and protein was measured by the bicinchoninic acid (BCA) method.
  • Total dry product measured after lyophilization was 3.28g for Porphyridium sp. and 2.Og for Porphyridium cruentum.
  • Total protein calculated as a percentage of total dry product was 12.6% for Porphyridium sp. and 15.0% for Porphyridium cruentum.
  • the first data set from sonication experiment 1 was obtained by subjecting the sample to sonication for a total time period of 60 minutes in 5 minute increments.
  • the second data set from sonication experiment 2 was obtained by subjecting the sample to sonication for a total time period of 6 minutes in 1 -minute increments.
  • Standard curves were generated using TFF-purif ⁇ ed, lyophilized, weighed, resuspended Porphyridium sp. exopolysaccharide.
  • the biomass was separated from the culture solution by centrifugation.
  • the centrifuge used was a Forma Scientific Centra-GP8R refrigerated centrifuge. The parameters used for centrifugation were 4200rpm, 8 minutes, rotor# 218. Following centrifugation, the biomass was washed with dH 2 ⁇ . The supernatant from the washings was discarded and the pelleted cell biomass was collected for the experiment.
  • a sample of lOO ⁇ L of the biomass suspension was collected at time point 0 (OTP) and suspended in 900 ⁇ L dH 2 O. The suspension was further diluted ten-fold and used for visual observation and DMMB assay.
  • the time point 0 sample represents the solvent- available polysaccharide concentration in the cell suspension before the cells were subjected to sonication. This was the baseline polysaccharide value for the experiments.
  • the container with the biomass was placed in an ice bath to prevent overheating and the ice was replenished as necessary.
  • the sample was prepared as follows for visual observation and DMMB assay: 100 ⁇ L of the biomass sample + 900 ⁇ L dH 2 Q was labeled as dilution 1. 100 ⁇ L of (i) dilution 1 + 900 ⁇ L dH ⁇ O for cell observation and DMMB assay.
  • EXAMPLE 4 Porphyridium sp. culture was centrifuged at 4000 rcf and supernatant was collected. The supernatant was divided into six 30ml aliquots. Three aliquots were autoclaved for 15 min at 121 0 C. After cooling to room temperature, one aliquot was mixed with methanol (58.3% vol/vol), one was mixed with ethanol (47.5% vol/vol) and one was mixed with isopropanol (50% vol/vol). The same concentrations of these alcohols were added to the three supernatant aliquots that were not autoclaved.
  • Polysaccharide precipitates from all six samples were collected immediately by centrifugation at 4000 rcf at 2O 0 C for 10 min and pellets were washed in 20% of their respective alcohols. Pellets were then dried by lyophilization and resuspended in 15 ml deionized water by placement in a 60 0 C water bath. Polysaccharide pellets from non-autoclaved samples were partially soluble or insoluble. Polysaccharide pellets from autoclaved ethanol and methanol precipitation were partially soluble. The polysaccharide pellet obtained from isopropanol precipitation of the autoclaved supernatant was completely soluble in water.
  • Monosaccharide analysis was performed by combined gas chromatography/mass spectrometry (GC/MS) of the per-O-trimethylsilyl (TMS) derivatives of the monosaccharide methyl glycosides produced from the sample by acidic methanolysis.
  • GC/MS gas chromatography/mass spectrometry
  • Methyl glycosides prepared from 500 ⁇ g of the dry sample provided by the client by methanolysis in 1 M HCl in methanol at 80°C (18-22 hours), followed by re-N-acetylation with pyridine and acetic anhydride in methanol (for detection of amino sugars). The samples were then per-O-trimethylsilylated by treatment with Tri-Sil (Pierce) at 80 0 C (30 mins). These procedures were carried out as previously described described in Merkle and Poppe (1994) Methods Enzymol. 230: 1-15; York, et al. (1985) Methods Enzymol. 118:3- 40. GC/MS analysis of the TMS methyl glycosides was performed on an HP 5890 GC interfaced to a 5970 MSD, using a Supelco DB-I fused silica capillary column (30m 0.25 mm ID).
  • Glucuronic acid n.d. n.d.
  • NANA N-acetyl neuraminic acid
  • Porphyridium sp. was grown as described. 2 liters of centrifuged Porphyridium sp. culture supernatant were autoclaved at 121°C for 20 minutes and then treated with 50% • isopropanol to precipitate polysaccharides. Prior to autoclaving the 2 liters of supernatant contained 90.38 mg polysaccharide. The pellet was washed with 20% isopropanol and dried by lyophilization. The dried material was resuspended in deionized water. The resuspended polysaccharide solution was dialyzed to completion against deionized water in a Spectra/Por cellulose ester dialysis membrane (25,000 MWCO). 4.24% of the solid content in the solution was proteins as measured by the BCA assay.
  • Porphyridium sp. was grown as described. 1 liters of centrifuged Porphyridium sp. culture supernatant was autoclaved at 121 0 C for 15 minutes and then treated with 10% protease (Sigma catalog number P-5147; protease treatment amount relative to protein content of the supernatant as determined by BCA assay). The protease reaction proceeded for 4 days at 37°C. The solution was then subjected to tangential flow filtration in a Millipore Pellicon ® cassette system using a 0.1 micrometer regenerated cellulose membrane. The retentate was diafiltered to completion with deionized water. No protein was detected in the diafiltered retentate by the BCA assay. See Figure 8.
  • the retentate can be autoclaved to achieve sterility if the filtration system is not sterile.
  • the sterile retentate can be mixed with pharmaceutically acceptable carrier(s) and filled in vials for injection.
  • the protein free polysaccharide can be fragmented by, for example, sonication to reduce viscosity for parenteral injection as, for example, an antiviral compound.
  • the sterile polysaccharide is not fragmented when prepared for injection as a joint lubricant.
  • Porphyridium sp. (UTEX 637) and Porphyridium cruentum (strain UTEX 161) were grown, to a density of 4 X 10 6 cells/mL, as described in Example 1. For each strain, about 2 X 10 6 cells/mL cells per well (-500 uL) were transferred to 11 wells of a 24 well microliter plate. These wells contained ATCC 1495 media supplemented with varying concentration of glycerol as follows: 0%, 0.1%, 0.25%, 0.5%, 0.75%, 1%, 2 %, 3%, 5%, 7% and 10%.
  • Duplicate microtiter plates were shaken (a) under continuous illumination of approximately 2400 lux as measured by a VWR Traceable light meter (cat # 21800-014), and (b) in the absence of light. After 5 days, the effect of increasing concentrations of glycerol on the growth rate of these two species of Porphyridium in the light was monitored using a hemocytometer. The results are given in Figure 3 and indicate that in light, 0.25 to 0.75 percent glycerol supports the highest growth rate, with an apparent optimum concentration of 0.5%. Cells in the dark were observed after about 2 weeks of growth. The results are given in Figure 4 and indicate that in complete darkness, 5.0 to 7.0% glycerol supports the highest growth rate, with an apparent optimum concentration of 7.0%.
  • Porphyridium sp. (UTEX 637) was grown to a density of approximately 4 X 10 6 cells/mL, as described in Example 1. Approximately 50 grams of wet pelleted, and washed cells were completely homogenized using approximately 20 minutes of sonication as described.
  • the homogenized biomass was mixed with carriers including, water, butylene glycol, mineral oil, petrolatum, glycerin, cetyl alcohol, propylene glycol dicaprylate/dicaprate, PEG-40 stearate, Cl 1-13 isoparaffin, glyceryl stearate, tri (PPG-3 myristyl ether) citrate, emulsifying wax, dimethicone, DMDM hydantoin, methylparaben, carbomer 940, ethylparaben, propylparaben, titanium dioxide, disodium EDTA, sodium hydroxide, butylparaben, and xanthan gum.
  • the mixture was then further homogenized to form a composition suitable for topical administration.
  • the composition was applied to human skin daily for a period of one week.
  • EXAMPLE 11 Approximately 4500 cells (300ul of 1,5x10 5 cells per ml) of P 'orphyridium sp. and Porphyridium omentum cultures in liquid ATCC 1495 ASW media were plated onto ATCC 1495 ASW agar plates (1.5% agar). The plates contained varying amounts of zeocin, sulfometuron, hygromycin and spectinomycin. The plates were put under constant artificial fluorescent light of approximately 480 lux. After 14 days, plates were checked for growth.
  • PIasmid pBluescript KS+ is used as a recipient vector for an expression cassette shown in SEQ ID NO:68.
  • the Porphyridium glycoprotein promoter is cloned into pBluescript KS+, followed by a cauliflower mosaic virus 3' UTR. Unique restriction sites are left between the promoter and 3'UTR.
  • a nucleic acid encoding the glucose transporter of SEQ ID NO:20 using preferred codons of Porphyridium sp. is cloned into the unique restriction sites between the promoter and 3'UTR.
  • the plasmid is used to transform Porphyridium sp. cells using the biolistic transformation parameters described in Plant Physiol. 2002 May;129(l):7-12. After transformation, some plated cells are scraped from the plate using a sterile cell scraper are transferred into Erlenmeyer flasks wrapped with aluminum foil sufficient to prevent the entry of light into the culture. Identical preparations of transformed, scraped cells are cultured, shaking at -50 rpm in 24 well plates in the dark, in ATCC 1495 media in the presence of 0.1, 1.0, and 2.5% glucose, and monitored for growth. Other cells are transformed on plates containing solid agar ATCC 1495 media, supplemented with either 0.1. 1.0, or 2.5% glucose, and monitored for growth in complete darkness.
  • EXAMPLE 13 Cultures of P 'orphyridium sp. (UTEX 637) and Porphyridium cruentum (strain UTEX 161) were subjected to chemical mutagenesis (from the protocol in Gorman DS, Levine RP. (1965) Proc Natl Acad Sci U S A. 54(6): 1665-9.). Cells were grown to a density of 4 X 10 6 cells/mL as described in Example 1. Cells were harvested, washed with 70 mM potassium phosphate buffer (pH 6.9) and resuspended to a density of 4 X 10 7 cells/mL.
  • EMS ethyl methane sulfonate
  • the cells were resuspended in 200 uL of ATCC 1495 media, and plated at three different concentrations (IX, 10 '2 X, 10 "4 X) on duplicate plates of ATCC 1495 media, and incubated under continuous light.
  • plated cells are scraped from the plate using a sterile cell scraper are transferred into Erlenmeyer flasks wrapped with aluminum foil sufficient to prevent the entry of light into the culture.
  • Identical preparations of transformed, scraped cells are cultured, shaking at ⁇ 50 rpm in 24 well plates in the dark, in ATCC 1495 media in the presence of 0.1 , 1.0, and 2.5% glucose, and monitored for growth.
  • Other cells are transformed on plates containing solid agar ATCC 1495 media, with either 0.1, 1.0, or 2.5% glucose, and monitored for growth in complete darkness.
  • Cell treated as described can also be cultured in the presence of an exogenous carbon source from Tables 2 or 3.
  • Cells prepared as described in Example 12 containing a monosaccharide transporter and capable of importing glucose, are cultured in ATCC 1495 media in the light in the presence of 1.0% glucose for approximately 12 days.
  • Exopolysaccharide is purified as described in Example 2.
  • Monosaccharide analysis is performed as described in Example 5.
  • Cells prepared as described in Example 12 containing a monosaccharide transporter and capable of importing xylose are cultured in ATCC 1495 media in the light in the presence of 1.0% xylose for approximately 12 days.
  • Exopolysaccharide is purified as described in Example 2.
  • Monosaccharide analysis is performed as described in Example 5.
  • Cells prepared as described in Example 12 containing a monosaccharide transporter and capable of importing galactose are cultured in ATCC 1495 media in the light in the presence of 1.0% galactose for approximately 12 days.
  • Exopolysaccharide is purified as described in Example 2.
  • Monosaccharide analysis is performed as described in Example 5.
  • Example 12 Cells prepared as described in Example 12 containing a monosaccharide transporter and capable of importing glucuronic acid, are cultured in ATCC 1495 media in the light in the presence of 1.0% glucuronic acid for approximately 12 days. Exopolysaccharide is purified as described in Example 2. Monosaccharide analysis is performed as described in Example 5.
  • Example 12 Cells prepared as described in Example 12 containing a monosaccharide transporter and capable of importing glucose, are cultured in ATCC 1495 media in the dark in the presence of 1.0% glucose for approximately 12 days. Exopolysaccharide is purified as described in Example 2. Monosaccharide analysis is performed as described in Example 5.
  • Example 12 Cells prepared as described in Example 12 containing a monosaccharide transporter and capable of importing xylose, are cultured in ATCC 1495 media in the dark in the presence of 1.0% xylose for approximately 12 days. Exopolysaccharide is purified as described in Example 2. Monosaccharide analysis is performed as described in Example 5.
  • Example 12 Cells prepared as described in Example 12 containing a monosaccharide transporter and capable of importing galactose, are cultured in ATCC 1495 media in the dark in the presence of 1.0% galactose for approximately 12 days. Exopolysaccharide is purified as described in Example 2. Monosaccharide analysis is performed as described in Example 5.
  • Example 12 Cells prepared as described in Example 12 containing a monosaccharide transporter and capable of importing glucuronic acid, are cultured in ATCC 1495 media in the dark in the presence of 1.0% glucuronic acid for approximately 12 days. Exopolysaccharide is purified as described in Example 2. Monosaccharide analysis is performed as described in Example 5.
  • 128mg of intact lyophilized Porphyridium sp. cells were ground with a mortar/pestle. The sample placed in the mortar pestle was ground for 5 minutes. 9.0 mg of the sample of the ground cells was placed in a micro centrifuge tube and suspended in lOOO ⁇ L of dH2O. The sample was vortexed to suspend the cells. A second sample of 9.0mg of intact, lyophilized Porphyridium sp. cells was placed in a micro centrifuge tube and suspended in 1000 ⁇ L of dH2O. The sample was vortexed to suspend the cells.
  • the suspensions of both cells were diluted 1:10 and polysaccharide concentration of the diluted samples was measured by DMMB assay. Upon grinding, the suspension of ground cells resulted in an approximately 10-fold increase in the solvent-accesible polysaccharide as measured by DMMB assay over the same quantity of intact cells.
  • Reduction in the particle size of the lyophilized biomass by homogenization in a mortar/pestle results in better suspension and increase in the solvent-accesible polysaccharide concentration of the cell suspension.
  • Porphyridium cruentum was grown as described above in ATCC 1495 media. Porphyridium cruentum culture supernatant were autoclaved at 121 0 C for 20 minutes. 1.333 liters of isopropanol was added to a 4 liter preparation of autoclaved supernatant to a concentration of 25% (vol/vol). Precipitated exopolysaccharide was removed. Additional isopropanol (381 mL, 786 mL, 167 mL, and 1.333 liters) was added stepwise to the preparation to produce (vol/vol) concentrations of isopropanol of 30%, 38.5%, 40%, and 50%, respectively.
  • Precipitated exopolysaccharide was removed after each increment of isopropanol was added. It was observed that very little additional exopolysaccharide was precipitated upon bringing the concentration from 38.5% to 40% and from 40% to 50%. It was also observed that significant amounts of salt were precipitated upon bringing the concentration from 38.5% to 40% and from 40% to 50%.
  • EXAMPLE 17 Preparation of DNA templates for Inverse PCR: Porphyridium sp. genomic DNA (40 ⁇ g) was digested with Sad at 37°C for 16h in a total volume of 200 ⁇ l. The digested DNA was extracted with once an equal volume of phenol : chloroform isoamyl alcohol (PCI, 25:24:1) followed by one extraction with equal volume of chloroform. The DNA was precipitated with 1/lOth volume of 3M sodium acetate, pH5.5 and 2.5 volumes of 100% ethanol. The DNA was pelleted by centrifugation, air dried and resuspended in 50 ⁇ l distilled water to a concentration of 250 ng/ ul.
  • PCI chloroform isoamyl alcohol
  • PCR was carried out in 25 ⁇ l volume using EXL polymerase (Stratagene, La JoIIa, CA) in 0.2ml thin-walled tubes. PCR reactions were carried out in two rounds. The first round of PCR was set up according to the manufacturer's instructions. The reaction contained 4% DMSO 5 1 ⁇ L stablilizing solution and 0.25 ⁇ M of primers 1 and 2 (primer #1, SEQ ID NO:49, primer #2, SEQ ID NO:50). Inverse PCR primers 1 and 2 were designed in opposite orientation to that of normal PCR and were designed to anneal to the Porphyridium sp. glycoprotein cDNA sequence (GenBank Accession number AY778963).
  • PCR cycle conditions were as follows: initial denaturation of 94°C for 2 minutes followed by 35 cycles of 92°C for 10 seconds, 60°C for 30 seconds and 68°C for 15 minutes, followed by a final extension at 68°C for 5 minutes.
  • the first round PCR product was diluted 100 fold, and 1 ⁇ L of the dilution was used as a template for a second round PCR.
  • Second round PCR was done using nested primers 3 and 4 (primer #3, SEQ ID NO:51, primer #4, SEQ ID NO:52).
  • PCR cycle conditions were as follows: initial denaturation of 94°C for 2 minutes followed by 35 cycles of 92°C for 10 seconds, 58°C for 30 seconds and 68°C for 6 minutes, followed by a final extension at 68°C for 5 minutes.
  • the 1.8kB promoter fragment was amplified by PCR and cloned into operable linkage with a zeocin resistance gene and the carbohydrate transporter cDNAs encoding the proteins of SEQ ID NOs:20, 22, 24, and 46-48 to generate 7 unique Porphyridium transformation vectors.
  • the promoter of SEQ ID NO:55 was cloned into operable linkage with a zeocin resitance cDNA encoding the protein of SEQ ID NO: 11.
  • the zeocin resistance cDNA was in operable linkage with the CMV 3' UTR of PCT application WO2006013572.
  • the plasmid was linearized by a restriction enzyme that did not cut in the promoter, cDNA, or 3'UTR.
  • Porphyridium sp. (strain UTEX 637) culture was grown in ATCC 1495 medium on a gyratory shaker under continuous light at 75 ⁇ mol photons m "2 sec "1 until it reached a cell density of 2xl0 6 cells/mL, following which they were incubated in the dark for 24 hours.
  • the dark adapted cells were harvested, and washed twice with sterile distilled water, and resuspended in a Tris-phosphate buffer (2OmM Tris-HCl, pH 7.0; 1 mM potassium phosphate) containing 50 mM sucrose to a density of 4xl0 8 cells/mL.
  • the cuvette was then incubated at room temperature for 5 minutes, following which the cell suspension was transferred to 50 mL of ATCC 1495 media, and shaken on a gyratory shaker for 2 days. Following recovery, the cells were harvested at low speed (4000 rpm), resuspended in distilled water and plated out at low density on solid media (ATCC 1495 + 1% agar) supplemented with 30 ⁇ g/mL zeocin (Invitrogen, Carlsbad, CA). The plates were incubated under continuous light at 75 ⁇ mol photons m "2 sec "1 . Transformants appeared as colonies in 2-3 weeks. EXAMPLE 19 Animal model
  • the total fat content of the control diet was 5% fed in the form of a mix of lard and safflower oil to provide a P/S ratio of 0.4.
  • the control diet alone will be fed to Group 1.
  • Figure 10 (a) Biomass and polysaccharide was matrixed into the oil component of the diets. Food intake and body weight of individual animals was monitored regularly through the feeding period.
  • Quadro ground biomass was homogenized in a Quadro FlO rotor homogenizer (Quadro Engineering Inc., Waterloo, Ontario, Canada).
  • Quadro FlO rotor homogenizer Quadro Engineering Inc., Waterloo, Ontario, Canada.
  • samples were cryogenically ball milled in a planetary ball mill (Retsch, PMlOO) at 10-80 g batch size. The powder was placed in a grinding bowl with eight to ten %-inch- diameter stainless steel balls. The sample was cooled with liquid nitrogen repeatedly. The material was milled at 400-550 rpm for 30 to 60 min. The final product was dried in a desiccator overnight.
  • hamsters in each group will be anaethesized with halothane and blood samples were collected and stored at -80 0 C for determination of total cholesterol, low density lipoprotein (LDL), and high density lipoprotein (HDL) subclass cholesterol and triglyceride (TG) levels. Measures of antioxidant status were also determined through TBARs assays. Lipid analysis
  • Plasma total cholesterol was measured using a VG Autoanalyzer in conjunction with commercial enzymatic kits and appropriate standards (Abbott Diagnostics, Montreal, Quebec). All samples were processed through the above system in duplicate.
  • Dvir et al. reported only a 21.8% reduction in TSC by feeding rodents microalgal biomass at 19% of the total diet. This is in contrast to the 5% of the total diet used in Group 3 and the 10% of the total diet in Group 5 of the instant experiment, which produced reductions of 35.9% and 54.2%, respectively.
  • the results from the instant experiment are also unexpected relative to the report by Ginzberg et al., where 5% and 10% diets only produced 11% and 28% reductions in TSC, respectively.
  • the results from Group 7 (62% reduction) also indicate the presence of unexpected synergy in a combination of cell homogenate and an additional agent such as phyto sterol.
  • Body weight was measured once a week and food consumption once every two days by weighing the difference of food cups before and after the feeding. Measurements of body composition were performed using DEXA analyses at sacrifice. Methods to determine body composition and energy expenditure (metabolism) were performed essentially as described by Lei et al. ("Relationship of total body fatness and five anthropometric indices in Chinese aged 20-40 years: different effects of age and gender" Eur. J. Clin. Nutri. (2006) 60:511-518) and Nagao et al. (“The lOtrans, 12 cis isomer of conjugated linoleic acid promotes energy metabolism in OLETF rats" Nutri. (2003) 19:652-656). Increasing satiety
  • compositions provided herein are useful, for example, in alleviating age-associated cognitive and motor changes in mammals. See for example Brain Res. 1995 Sep 25;693(l-2):88-94.
  • Porphyridium sp. biomass homogenate was tested for COXl and COX2 inhibition activity.
  • Porphyridium sp. biomass was grown as described in Example 1 , recovered by centrifugation, washed with deionizer water to remove salts, and centrifugd again to form a pelleted paste. Nitrogen was bubbled through the paste for 30 minutes to displace dissolved oxygen and minimize subsequent oxidation. The paste was then passed through a model 11 OY Microfluidics Microfluidizer ® at 22,000 PSI with cooling, and the process repeated until at least 50% of the cells were broken as determined by microscopic examination. Nitrogen was once again bubbled through the paste, which wais then lyophilized after shell freezing in dry ice ethanol. The dried cell mass was then ground to a fine powder with a Braun ® kitchen homogenizer.
  • the powder was analyzed for inhibition of constitutive cyclooxygenase (COXl) and inducible cyclooxygenase (COX2) at 37 0 C by monitoring oxygen consumption using an Oxytherm Electrode Unit by Hansatech.
  • the ICs 0 values are equal to the concentration of the sample that inhibits 50% activity of the cyclooxygenase in the reaction mixture. The results are shown in Table 14.
  • Results indicate surprisingly significant anti-inflammatory activity, particulary for a natural product.
  • Certain aspects of the invention are based upon the discovery of an antiinflammatory effect in animals that ingest the cell material (biomass, homogenate, and/or polysaccharides) described herein. While the basis of the anti-inflammatory effect has yet to be completely elucidated, the materials of the invention both increase the antioxidant status of mammalian plasma (see Example 23 below) and inhibit cyclooxygenase activity.
  • the cell material has anti -inflammatory activity demonstrated through multiple methods of analysis and shown to be exerted through independent meachansism that would not have been expected.
  • Two filter-purified polysaccharide preparations (containing 3.57% and 4.23% sulfur by weight), prepared as described in Example 24, were subjected to oxygen radical absorption capacity (ORAC) analysis as described in Journal of Agricultural and Food Chemistry.; 2001 ;49(10); 4619-4626 and Journal of Agricultural and Food Chemistry.; 2002, 50(7); 1815-1821 ,
  • ORAC oxygen radical absorption capacity
  • the polysaccharide containing 3.57% sulfur by weight did not exhibit ORAC activity.
  • the polysaccharide containing 4.23% sulfur by weight exhibited ORAC activity of 2.0 trolox equivalents per gram.
  • Porphyridium cruentum and Porphyridium sp. were grown in artificial seawater media essentially as described in Example 1 except that the amount OfMgSO 4 was varied.
  • Porphyridium sp.cells were grown in 17mM MgSO 4 .
  • Porphyridium cruentum was grown in 12OmM, 60OmM, 750 mM, IM 3 and 2M MgSO 4 . Cell division occurred at all concentrations.
  • Polysaccharide was purified essentially as described in Example 2 from the 120 and 60OmM cultures, to the point where all soluble protein and small molecules were removed. Sulfur content was analyzed according to US EPA SW846, Method 6010B, Inductively Coupled Plasma- Atomic Emission Spectrometry.
  • the polysaccharide purified from the 17, 120 and 60OmM cultures contained 3.57, 4.23 and 5.57% sulfur, respectively. As disclosed herein, these results reflect an unexpected discovery because the skilled person would not expect that the microalgae would survive in media containing more than 100 mM sulfate.
  • the CMV 3'UTR was also in operable linkage with the ble cDNA.
  • the plasmid was linearized by Kpnl,. which does cut in the promoter, ble cDNA, or 3'UTR, prior to transformation.
  • the biolistic particle delivery system PDS 1000/ He (Bio-Rad, USA) was used for transformation. Porphyridium sp. culture was grown to logarithmic phase ( ⁇ 2 x 10 6 cells/mL) in liquid ATCC 1495 media under continuous light (approximately 75 umol/photons/m 2 ). Cells from this culture were harvested at 4,000 rpm at room temperature. The cell pellet was washed twice with sterile distilled water. Cells were resuspended in fresh ATCC 1495 media to the same cell density i.e.. ⁇ 2 x 10 6 cells/mL and incubated in the dark for 16 hours.
  • the dark adapted cells were then harvested at 4000 rpm at room temperature, resuspended in fresh ATCC 1495 media to a density of ⁇ 2 x 10 8 cells/mL. Approximately 1 x 10 s cells were transferred to each ATCC 1495 agarose plate. Filter sterilized DNA from the plasmids was coated onto 550 nm gold particles (catalog number S04e, Seashell Technology, USA) according to the manufacturer's protocol. For each of the particle bombardments, 1 ug of plasmid DNA was used.
  • the negative controls were bombarded in identical fashion with gold particles coated with a plasmid containing the Porphyridum glycoprotein promoter, SEQ ID NO:44, and the CMV 3'UTR, (SEQ ID NO:69), with no zeocin resistance gene.
  • Each of the particle bombardments were performed using 1350 psi rupture disks, at bombardment distance of 9 cm, and under 28 in. Hg vacuum.
  • the bombarded cells were scraped off the plates, and transferred to 100 ml of fresh ATCC 1495 media, and shaken under continuous light (approximately 75 umol/ m 2 ) for 3 days.
  • the cells were harvested at 4,000 rpm at room temperature, and plated onto ATCC 1495 plates supplemented with 30 ug/mL Zeocin (Invitrogen, Carlsbad, CA, USA) at a cell density of 1 x 10 7 cells/plate. These plates were incubated under light (approximately 25 umol/ m 2 ) for 4-5 weeks. Zeocin resistant colonies growing on these plates were scored as transformants and transferred onto fresh ATCC 1495 plates supplemented with 30 ug/mL Zeocin (Invitrogen, Carlsbad, CA, USA) for growth and analysis.
  • Zeocin Invitrogen, Carlsbad, CA, USA
  • Genotyping was performed on genomic DNA extracted from wild type, transformant#l, and transformant#2 DNA with plasmid DNA used as a template positive control and water in place of DNA as a template negative control.
  • a segment of the Porphyridium glycoprotein (GLP) gene promoter was used as a target positive control.
  • the following primer sets were used for the genotyping PCR: BIe- FWD (SEQ ID NO: 62) and BIe-REV (SEQ ID NO: 63), GLP-FWD (SEQ ID NO: 64) and (SEQ ID NO: 65), GLUTl- FWD (SEQ ID NO: 66) and GLUTl-REV (SEQ ID NO: 67).
  • PCR profile used was as follows: 94°C denaturation for 5 min; 35 cycles of 94°C for 30 sec, 51 0 C or 6O 0 C (51 0 C for glycoprotein gene & GLUTl and 6O 0 C for ble) for 30 sec, 72°G for 2 min; 72 0 C for 5 min. Results are shown in Figure 15.
  • Figure 15 (a) demonstrates that the ble gene was present in both transformants, as the expected 300bp product was generated.
  • Figure 15(b) demonstrates that the genomic DNA extraction and amplification was working, as the expected 948bp glycoprotein promoter fragment was generated.
  • Figure 15(c) demonstrates that the GLUTl gene was present transformant #1, as the expected 325bp product was generated.
  • DNA ladder was from BioNexus, Inc., All Purpose Hi-Lo DNA Marker, Catalog No: BN2050.
  • plasmids that have not been linearized can be maintained as episomes for a period of time before being degraded and can serve as template during PCR despite not having been integrated into a chromosome of a host organism.
  • microalgal strains may genotype positive despite the absence of stable chromosomal integration of the vector.
  • Antibiotic resistant strains are known to arise due to mutagenesis caused by chromosomal damage from biolistic particles, electroporation conditions, and random genetic variation that is known to occur in microbial organisms. Southern blot analysis was performed to conclusively confirm the integration of the GLUTl construct into the genome of transformant #1.
  • Southern blot analysis was performed on transformant #1. 20 ⁇ g genomic DNA from wild type and transformant#l were individually digested with Hinc II, Sac I , Xho I and separated on a 1% agarose gel. DNA was transferred onto Nylon membrane (Hybond N+, Amersham Biosciences). A 1495 bp fragment containing the entire coding region of the GLUTl gene was used as a probe. DIG labeled probes were generated for each probe fragment using the DIG High Prime DNA labeling and detection Kit according to the manufacturers instructions (Roche). The hybridizing bands were detected using the colorimetric detection reagents provided by the manufacturer.
  • Figure 16 demonstrates that the GLUTl plasmid was stably integrated into the genome of transformant#l while no detectable signal arose from wild type genomic DNA.
  • the specific band was in a different position for each different restriction enzyme used to digest the genomic DNA.
  • This data conclusively demonstrates a species of the genus Porphyridium containing an exogenous gene encoding a carbohydrate transporter integrated into a chromosome of the organism.
  • the carbohydrate transporter gene is in operable linkage with a promoter endogenous to a species of the genus Porphyridium.
  • the carbohydrate transporter gene is in operable linkage with a promoter endogenous to a Rhodophyte.
  • Biotinylated hyaluronic acid was covalently attached to the wells of a 96-well plate. Samples containing hyaluronidase and various test materials were then added to the wells. The hyaluronidase degrades the bound hyaluronic acid, resulting in a decrease in the amount of biotin covalently linked to the well plate. At the end of the incubation period the reaction was stopped and the well plate was was washed to remove the hyaluronidase. The remaining bHA was detected using an avidin bound peroxidase enzyme. When an appropriate substrate is added, the peroxidase enzyme generated a color signal in proportion to the amount of bHA.
  • the color signal was measured spectrophotometrically, and was inversely proportional to the amount of hyaluronidase activity in the sample.
  • materials that inhibited hyaluronidase resulted in a greater color signal, since more of the bHA remained intact.
  • Frost, G., L, Stern, R. A Microtiter-Based Assay for Hyaluronidase Activity Not Requiring Specialized Reagents. Analytical Biochemistry 251, 263-269: 1997.
  • Test Material A was supplied as a powder type material. For this study, 100 mg of this material was combined with either 5 ml of ethanol or 5 ml of ultrapure water in 15 ml centrifuge tubes. After combining, the mixtures were vortexed, then placed onto a rocking platform for approximately 30 minutes at room temperature, and then centrifuged at 1,000 RPM for 5 minutes. The supernatants were then used at the final concentrations listed in the results section. Test Material B was supplied as a thick, viscous solutions (3%).
  • Anti -Hyaluronidase Assay Immobilization of bHA onto 96-well plates
  • a solution of sulfo-NHS (0.184 mg/ml) and bHA (0.2 mg/ml) was prepared in distilled water. 50 ⁇ l of this solution was then added to the wells of a 96-well Covalink-NH plate.
  • a solution of EDC (0.123 mg/ml) was then prepared in distilled water and 50 ⁇ l of this solution was added to each well (which resulted in a final concentration of 10 ⁇ g/well bHA and 6.15 ⁇ g/well of EDC).
  • the well plate was then incubated overnight at 4 ⁇ 2°C or for 2 hours at room temperature. After the incubation the plate was washed three times with PBS containing 2 M NaCl and 50 mM MgSO 4 .
  • the well plate Prior to the assay, the well plate was equilibrated with assay buffer (0.1 M formate [pH 4.5], 0.1 M NaCl, 1% Triton X-IOO 3 5 mM saccharolactone).
  • assay buffer 0.1 M formate [pH 4.5], 0.1 M NaCl, 1% Triton X-IOO 3 5 mM saccharolactone.
  • the test materials were prepared in assay buffer at 2x their final concentration (heparin was used as a positive control, 1 mg/ml final concentration).
  • 50 ⁇ l of each of the prepared test materials was added to three wells on the well plate, followed by the addition of 50 ⁇ l of assay buffer containing hyaluronidase will be added to each well (0.1 mg/ml).
  • an avidin/biotin-peroxidase complex was prepared in 10.5 ml of PBS containing 0.1% Tween 20 using an ABC kit. This mixture was incubated for at least 30 minutes prior to use. After the plate was washed, 100 ⁇ l of the avidin/biotin-peroxidase solution was added to each well and the plate was incubated for another 30 minutes at room temperature. The plate was washed three times with PBS containing 2 M NaCl, 50 mM MgSO 4 and 0.05% Tween 20.
  • substrate solution one 10 mg tablet of OPD in 10 ml of 0.1 M citrate-PO 4 buffer supplemented with 7.5 ⁇ l of 30% H 2 O 2 .
  • substrate solution 100 ⁇ l of substrate solution (one 10 mg tablet of OPD in 10 ml of 0.1 M citrate-PO 4 buffer supplemented with 7.5 ⁇ l of 30% H 2 O 2 ) was added to each well.
  • the plate was incubated in the dark for 10-20 minutes and then read at 460 ran using a plate reader.
  • the substrate solution was also added to three wells that were not treated with test materials or the avidin/biotin-peroxidase solution and were used as a blank for the absorbance measurements.
  • MATERIAL A Porphyridium sp. biomass homogenized (Quadro FlO); cells grown as described in Example 1
  • Test Material Identification 3% MATERIAL B: Exopolysaccahride from Porphyridium sp. purified as described in Example 2

Abstract

Provided herein are polysaccharide compositions and methods of culturing microalgae to produce polysaccharides. Also provided are methods of using polysaccharides for applications such as reducing cholesterol in mammals, inactivating viruses, stabilizing foods, and other uses. Also provided are transgenic algae capable of utilizing fixed carbon sources for energy. Also provided herein are novel nucleic acid sequences from red microalgae.

Description

Nutraceutical Compositions from Microalgae and Related Methods of Production and
Administration
BACKGROUND OF THE INVENTION
Carbohydrates have the general molecular formula CH2O, and thus were once thought to represent "hydrated carbon". However, the arrangement of atoms in carbohydrates has little to do with water molecules. Starch and cellulose are two common carbohydrates. Both are macromolecules with molecular weights in the hundreds of thousands. Both are polymers; that is, each is built from repeating units, monomers, much as a chain is built from its links.
Three common sugars share the same molecular formula: CeH^Oό- Because of their six carbon atoms, each is a hexose. Glucose is the immediate source of energy for cellular respiration. Galactose is a sugar in milk. Fructose is a sugar found in honey. Although all three share the same molecular formula (CβHπOδ), the arrangement of atoms differs in each case. Substances such as these three, which have identical molecular formulas but different structural formulas, are known as structural isomers. Glucose, galactose, and fructose are "single" sugars or monosaccharides.
Two monosaccharides can be linked together to form a "double" sugar or disaccharide. Three common disaccharides are sucrose, common table sugar (glucose + fructose); lactose, the major sugar in milk (glucose + galactose); and maltose, the product of starch digestion (glucose + glucose). Although the process of linking the two monomers is complex, the end result in each case is the loss of a hydrogen atom (H) from one of the monosaccharides and a hydroxyl group (OH) from the other. The resulting linkage between the sugars is called a glycosidic bond. The molecular formula of each of these disaccharides is Ci2H22Oi i = 2 CeH^O6 - H2O. All sugars are very soluble in water because of their many hydroxyl groups. Although not as concentrated a fuel as fats, sugars are the most important source of energy for many cells.
BRIEF SUMMARY OF THE INVENTION
The present invention relates to polysaccharides from microalgae. Representative polysaccharides include those present in the cell wall of microalgae as well as secreted polysaccharides, or exopolysaccharides. In addition to the polysaccharides themselves, such as in an isolated, purified, or semi-purified form, the invention includes a variety of compositions containing one or more microalgal polysaccharides as disclosed herein. The compositions include nutraceutical, cosmeceutical, industrial and pharmaceutical compositions which may be used for a variety of indications and uses as described herein. Other compositions include those containing one or more microalgal polysaccharides and a suitable carrier or excipient for topical or oral administration.
The invention further relates to methods of producing or preparing microalgal polysaccharides. In some disclosed methods, exogenous sugars are incorporated into the polysaccharides to produce polysaccharides distinct from those present in microalgae that do not incorporate exogenous sugars. The invention also includes methods of trophic conversion and recombinant gene expression in microalgae. In some methods, recombinant microalgae are prepared to express heterologous gene products, such as mammalian proteins as a non-limiting example, while in other embodiments, the microalgae are modified to produce more of a small molecule already made by microalgae in the absence of genetic modification.
Additionally, the invention relates to methods of using the polysaccharides and/or compositions containing them. In some disclosed methods, one or more polysaccharides are used to lower cholesterol, prevent sexually transmitted diseases, lubricate joints, regulate insulin levels, enhance cosmetics, stabilize or emulsify foods, and treat or effect prophylaxis of inflammation.
So in one aspect, the invention includes a nutraceutical composition containing one or more polysaccharides disclosed herein and a carrier suitable for human consumption. In other aspects, the composition contains the carrier and homogenized microalgae cells, such as red microalgae cells as a non-limiting example. In some embodiments, the composition contains the carrier and a purified first polysaccharide produced from a microalgal species listed in Table 1, which lists non-limiting examples of microalgae for the practice of the invention. Non-limiting examples of the carrier include a human nutritional supplement, such as vitamins, minerals, herbal extracts, monosaccharides or polysaccharides (e.g. glucosamine, glucosamine sulfate, chondroitin, or chondroitin sulfate, etc.) and proteins (e.g. protein supplements, etc.); a human food product; and various human foods perse.
In another aspect, the invention relates to compositions for topical application. In some embodiments, the composition is that of a cosmeceutical. A cosmeceutical may contain one or more microalgal polysaccharides, or a microalgal cell homogenate, and a topical carrier. In some embodiments, the carrier may be any carrier suitable for topical application, such as, but not limited to, use on human skin or human mucosal tissue. In some embodiments, the composition, may contain a purified microalgal polysaccharide, such as an exopolysaccharide, and a topical carrier.
As a cosmeceutical, the composition may contain a microalgal polysaccharide or homogenate and other component material found in cosmetics. In some embodiments, the component material may be that of a fragrance, a colorant (e.g. black or red iron oxide, titanium dioxide and/or zinc oxide, etc.), a sunblock (e.g. titanium, zinc, etc.), and a mineral or metallic additive.
In other aspects, the invention includes methods of preparing or producing a microalgal polysaccharide. In some aspects relating to an exopolysaccharide, the invention includes methods that separate the exopolysaccharide from other molecules present in the medium used to culture exopolysaccharide producing microalgae. In some embodiments, separation includes removal of the microalgae from the culture medium containing the exopolysaccharide, after the microalgae has been cultured for a period of time. Of course the methods may be practiced with microalgal polysaccharides other than exopolysaccharides. In other embodiments, the methods include those where the microalgae was cultured in a bioreactor, optionally where a gas is infused into the bioreactor.
In one embodiment, the invention includes a method of producing an exopolysaccharide, wherein the method comprises culturing microalgae in a bioreactor, wherein gas is infused into the bioreactor, separating the microalgae from culture media, wherein the culture media contains the exopolysaccharide; and separating the exopolysaccharide from other molecules present in the culture media.
The microalgae of the invention may be that of any species, including those listed in Table 1 herein. In some embodiments, the microalgae is a red algae, such as the red algae Porphyridium, which has two known species {Porphyridium sp. and Porphyridium cruentum) that have been observed to secrete large amounts of polysaccharide into their surrounding growth media. In other embodiments, the microalgae is of a genus selected from Rhodella, Chlorella, and Λchnanthes. Non-limiting examples of species within a microalgal genus of the invention include Porphyridium sp., Porphyridium cruentum, Porphyridium purpureum, Porphyridium aerugineum, Rhodella maculata, Rhodella reticulata, Chlorella autotrophica, Chlorella stigmatophora, Chlorella capsulata, Achnanthes brevipes and Achnanthes longipes.
In some embodiments, a polysaccharide preparation method is practiced with culture media containing over 26.7, or over 27, rnM sulfate (or total SO4 2"). Non-limiting examples include media with more than about 28, more than about 30, more than about 35, more than about 40, more than about 45, more than about 50, more than about 55, more than about 60, more than about 65, more than about 70, more than about 75, more than about 80, more than about 85, more than about 90, more than about 95, or more than about 100 mM sulfate. In additional embodiments, more than about 150, more than about 200, more than about 250, more than about 300, more than about 350, more than about 400, more than about 450, more than about 500, more than about 550, more than about 600, more than about 650, more than about 700, or more than about 750, more than about 800, more than about 850, more than about 900, more than about 950, or more than about 1 M sulfate may be used to culture the microalgae and produce polysaccharides. Sulfate in the media may be provided in one or more of the following forms: Na2SO4-IO H2O, MgSO4-7H20, MnSO4, and CuSO4.
Other embodiments of the method include the separation of an exopolysaccharide from other molecules present in the culture media by tangential flow filtration. Alternatively, the methods may be practiced by separating an exopolysaccharide from other molecules present in the culture media by alcohol precipitation. Non-limiting examples of alcohols to use include ethanol, isopropanol, and methanol.
In other embodiments, a method may further comprise treating a polysaccharide or exopolysaccharide with a protease to degrade polypeptide (or proteinaceous) material attached to, or found with, the polysaccharide or exopolysaccharide. The methods may optionally comprise separating the polysaccharide or exopolysaccharide from proteins, peptides, and amino acids after protease treatment.
In addition to preparation or production of a polysaccharide per se, the invention includes methods of preparing a composition containing a microalgal polysaccharide or homogenate. In some embodiments, a method of producing a nutraceutical composition is described. As a non-limiting example;, the composition may be prepared by drying a homogenate of microalgae after the microalgae have been disrupted to produce a homogenate. In some embodiments, the microalgae is separated from the culture medium used to grow the microalgae. One non-limiting example of microalgae uses red microalgae to prepare the homogenate. Thus a homogenate processed as described herein may be combined with an appropriate carrier to form a nutraceutical of the invention.
In addition to a composition comprising a homogenate of red microalgae cells, the invention further includes a composition comprising a first component comprising cell material of the genus Porphyridium and at least one cholesterol lowering agent or compound. In some embodiments, the cell material is a cholesterol lowering homogenate, or a preparation of sulfated polysaccharides, and the agent or compound is selected from a plant phytosterol, a limonoid, a flavonoid, a tocotrienol, or any combination thereof. In additional embodiments, the agent is an inhibitor of HMG CoA reductase, such as a statin. The cell material may also be cells (biom&ss) per se, and in further embodiments, the cell material and cholesterol lowering agent may be packaged for sale as a single unit.
A method of formulating the above described composition is also disclosed. In some embodiments, the method comprises culturing cells of the genus Porphyridium; separating the cells from culture media to prepare a cell containing material (biomass); drying the cell containing material; and mixing the dried material with at least one cholesterol lowering agent or compound. In some embodiments, the dried material is homogenized before mixing with the agent or compound. In some embodiments the cells are homogenized before drying. In representative cases, the agent or compound is selected from a plant phytosterol, a limonoid, a flavonoid, a tocotrienol, or a combination thereof, including a combination of two, three or all four thereof.
In other embodiments, a method of formulating a cosmeceutical composition is disclosed. As one non-limiting example, the composition may be prepared by adding separated polysaccharides, or exopolysaccharides, to homogenized microalgal cells before, during, or after homogenization. Both the polysaccharides and the microalgal cells may be from a culture of microalgae cells in suspension and under conditions allowing or permitting cell division. The culture medium containing the polysaccharides is then separated from the microalgal cells followed by 1) separation of the polysaccharides from other molecules in the medium and 2) homogenization of the cells.
Other compositions of the invention may be formulated by subjecting a culture of microalgal cells and soluble exopolysaccharide to tangential flow filtration until the composition is substantially free of salts. Alternatively, a polysaccharide is prepared after proteolysis of polypeptides present with the polysaccharide. The polysaccharide and any contaminating polypeptides may be that of a culture medium separated from microalgal cells in a culture thereof. Ln some embodiments, the cells are of the genus Porphyridium.
In further aspects, the invention relates to methods of using a composition of the invention. In one aspect, a method of lowering serum cholesterol is described. The method may include orally administering, to a subject in need thereof, a polysaccharide produced by microalgae in combination with a biologically acceptable carrier. In other embodiments, such a method is practiced by using a cholesterol lowering composition as described herein. One non-limiting example of such a composition contains a purified microalgal exopolysaccharide, or a microalgal cell homogenate, and a carrier suitable for human oral consumption.
In another embodiment, a method of preventing a sexually transmitted disease is described. In one embodiment, a method includes administration of a solution comprising a polysaccharide produced by microalgae and use of a prophylactic device. In other embodiments, the solution is administered via the device.
In a further embodiment, a method of mammalian joint lubrication is described. In one embodiment, a method includes injecting polysaccharide produced by microalgae into a cavity containing synovial fluid.
In yet another embodiment, a method of regulating insulin is described. Tn one embodiment, a method includes administering a polysaccharide produced by microalgae.
In an additional embodiment, a method of cosmetic enhancement is described. In one embodiment, a method may include injecting a polysaccharide produced by microalgae into mammalian skin.
In a yet further embodiment, a method of stabilizing or emulsifying a food composition is described. In one embodiment, a method includes adding a polysaccharide produced by microalgae into a food composition.
In a yet additional embodiment, a method of treating or effecting prophylaxis of mammalian inflammation is described. In one embodiment, a method includes administering a polysaccharide produced by microalgae to a mammal.
An additional aspect of the invention includes a method of reducing reactive oxygen species (ROS) in a mammal. In some embodiments, the method comprises administering a composition comprising disrupted red microalgae cells as described herein. In other embodiments, the composition comprises one or more other agents or compounds with antioxidant activity. In further embodiments, the method may be used to lower the level of ROS, or reduce or decrease the amount of damage caused by ROS.
A composition comprising homogenized red microalgae cells as described herein may also be used in additional methods of the invention. In one set of embodiments, the composition may be used in a method of delaying the onset of, or treating, a neurodegenerative disease in a mammal. The neurodegenerative disease may be selected from Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, Freidreich's ataxia, Huntington's disease, or a Prion disease.
The composition may also be used in a method of reducing organ transplant rejection, a method of reducing the percentage of a mammalian body that is made of fat, a method of increasing satiety (or decreasing appetite) in a mammal, a method of increasing the energy expenditure of a mammal, a method of reducing the body weight of a mammal, a method of reducing inflammation in a mammal, or a method for the prevention and/or treatment of atherosclerosis. Each of these methods may comprise administering a composition comprising homogenized red microalgae cells in a sufficient or effective amount as described herein.
In further aspects, the invention describes recombinant methods to modify microalgal cells. In some embodiments, the methods produce a microalgal cell that expresses an exogenous gene product. The exogenous gene product may encode a carbohydrate transporter protein as a non-limiting example. In other embodiments, a microalgal cell containing an exogenous gene encoding a mammalian growth hormone is described. The recombinantly modified cells per se, whether newly created or maintained in culture, are also part of the invention.
The invention also describes methods of recombinantly modifying a microalgal cell. In some embodiments, a method of trophically converting a microalgal cell, such as members of the genus Porphyridium, is described. The method may include selecting cells for a phenotype after transforming cells with a nucleic acid molecule in an expressible form, hi some methods, the phenotype may be the ability to undergo cell division in the absence of light and/or in the presence of a carbohydrate that is transported by a carbohydrate transporter protein encoded by the nucleic acid molecule.
These methods may also be considered a method of expressing an exogenous gene in a microalgal cell. The method may include use of an expression vector containing a nucleic acid sequence encoding a polypeptide, such as a carbohydrate transporter protein. Alternatively, the method may include transforming a microalgal cell with a dual expression vector containing 1) a resistance cassette with a gene encoding a protein that confers resistance to an antibiotic, such as zeocin as a non-limiting example, operably linked to a promoter active in microalgae; and 2) a second expression cassette with a gene encoding a second protein operably linked to a promoter active in microalgae. After transformation, cells may be selected for the ability to survive in the presence of the antibiotic, such as at least 2.5 μg/ml zeocin as a non-limiting example where zeocin resistance is used. Alternatively, the antibiotic can be at least 3.0 μg/ml zeocin, at least 4.0 μg/ml zeocin, at least 5.0 μg/ml zeocin, at least 6.0 μg/ml zeocin, at least 7.0 μg/ml zeocin, and at least 8.0 μg/ml zeocin. The invention further relates to microalgal cells expressing a carbohydrate transporter protein for use in a method of producing a glycopolymer. In some embodiments, the method may include providing a transgenic cell containing an expressible gene encoding a monosaccharide transporter; and culruring the cell in the presence of at least one monosaccharide, transported into the cell by the transporter, wherein the monosaccharide is incorporated into a polysaccharide made by the cell.
Alternatively, a method of trophically converting a microalgae cell may include selecting for the ability to undergo cell division in the absence of light after subjecting the microalgal cell to a mutagen and placing the cell in the presence of a molecule listed in Tables 2 or 3 herein.
Another embodiment of the invention includes a recombinant nucleic acid comprising an endogenous promoter from Porphyridium sp. Also included in the invention is a recombinant promoter sequence that drives expression of the gene encoding the glycoprotein associated with the polysaccharide from Porphyridium sp., GenBank accession number AAV48590. In some embodiments the promoter or a segment thereof is operably linked to a coding region encoding a carbohydrate transporter or an antibiotic resistance gene.
The invention further comprises a cell of the genus Porphyridum, wherein the genome of the cell contains an exogenous gene encoding a carbohydrate transport protein. In some embodiments the species is selected from the group consisting of Porphyridium sp. UTEX 637 or a strain derived from Porphyridium sp. UTEX 637, Porphyridium cruentum UTEX 161 or a strain derived from Porphyridium cruentum UTEX 161, Porphyridium aerugineum or a strain derived from Porphyridium aerugineum, Porphyridium sordidum or a strain derived from Porphyridium sordidum, Porphyridium purpureum or a strain derived from Porphyridium purpureum. In some embodiments the carbohydrate transport protein has at least 60% amino acid identity with a member of the group consisting of SEQ ID NOs: 20, 22, 24, 26, 27, 29-39, and 46-48.
The details of additional embodiments of the invention are set forth in the accompanying drawings and the description below. Other features and advantages of the invention will be apparent from the drawings and detailed description, and from the claims. BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows precipitation of 4 liters of Porphyridium cruentum exopolysaccharide using 38.5% isopropanol. (a) supernatant; (b) addition of 38.5% isopropanol; (c) precipitated polysaccharide; (d) separating step.
Figure 2 shows Porphyridium sp. cultured on agar plates containing various concentrations of zeocin.
Figure 3 shows growth of Porphyridium sp. and Porphyridium cruentum cells grown in light in the presence of various concentrations of glycerol.
Figure 4 shows Porphyridium sp. cells grown in the dark in the presence of various concentrations of glycerol.
Figure 5 shows levels of solvent-accessible polysaccharide in Porphyridium sp. homogenates subjected to various amounts of physical disruption from Sonication Experiment 1.
Figure 6 shows levels of solvent-accessible polysaccharide in Porphyridium sp. homogenates subjected to various amounts of physical disruption from Sonication Experiment 2.
Figure 7 shows sexually transmitted disease prevention devices containing various amounts of exopolysaccharide.
Figure 8 shows protein concentration measurements of autoclaved, protease-treated, and diafiltered exopolysaccharide.
Figure 9 shows various amounts and ranges of amounts of compounds found per gram of cells in cells of the genus Porphyridium.
Figure 10 (a) shows the diets used for animals of Groups 1 to 8. Figure 10 (b) shows total serum cholesterol (TSC) levels (in mmol/L) in mice administered the diets of Groups 1 to 8. Figure 10 (c) shows the mean TSC levels in mice administered the diets of Groups 1 to 8.
Figure 11 (a) shows the average food intake (grams) per day in mice administered the diets of Groups 1 to 8. Figure 11 (b) shows the average body weight in mice administered the diets of Groups 1 to 8 over a 4 week period.
Figure 12 shows the percentage of total fat mass in mice administered the diets of Groups 1 to 8.
Figure 13 shows the energy expenditure in mice administered the diets of Groups 1 to 8. Figure 14 shows the mean concentration of plasma antioxidants in mice administered the diets of Groups 1 to 8.
Figure 15(a) shows PCR genotyping of two Porphyridiurn transformants for the ble antibiotic resistance transgene. Figure 15(b) shows PCR genotyping of two Porphyridium transformants for the endogenous glycoprotein gene promoter. Figure 15(c) shows PCR genotyping of a selected Porphyridium transformant for an exogenous gene encoding a recoded human GLUTl transporter.
Figure 16 shows a Southern blot indicating chromosomal integration of an exogenous gene encoding a recoded human GLUTl transporter.
DETAILED DESCRIPTION OF THE INVENTION
U.S. Patent 7,135,290 is hereby incorporated in its entirety for all purposes. This application is a continuation-in-part of and claims priority to U.S. Patent application No: 11/336,426, filed January 19, 2006, entitled "Polysaccharide Compositions and Methods of Administering, Producing, and Formulating Polysaccharide Compositions", which is hereby incorporated in its entirety for all purposes. This application is a continuation-in- part of and claims priority to U.S. Patent application No:l 1/337,103, filed January 19, 2006, entitled "Methods and Compositions for Improving the Health and Appearance of Skin", which is is hereby incorporated in its entirety for all purposes. This application is a continuation-in-part of and claims priority to U.S. Patent application No:l 1/336,656, filed January 19, 2006, entitled "Devices and Solutions for Prevention of Sexually Transmitted Diseases", which is hereby incorporated in its entirety for all purposes. This application is a continuation-in-part of and claims priority to U.S. Patent application No:l 1/336,428, filed January 19, 2006, entitled "Methods and Compositions for Cholesterol Reduction in Mammals", which is hereby incorporated in its entirety for all purposes. This application is a continuation-in-part of and claims priority to U.S. Patent application No: 11/337,171, filed January 19, 2006, entitled "Methods and Compositions for Reducing Inflammation and Preventing Oxidative Damage", which is hereby incorporated in its entirety for all purposes. This application is a continuation-in-part of and claims priority to U.S. Patent application No: 11/336,431, filed January 19, 2006, entitled "Methods and Compositions for Thickening, Stabilizing and Emulsifying Foods", which is hereby incorporated in its entirety for all purposes. This application is a continuation-in-part of and claims priority to U.S. Patent application No: 11/336,430, filed January 19, 2006, entitled "Methods and Compositions for Joint Lubrication", which is hereby incorporated in its entirety for all purposes. This application is a nonprovisional of and claims priority to U.S. Patent application No: 60/832,091, filed July 20, 2006, entitled "Decolorized Microalgal Compositions for Skin Care Products", which is hereby incorporated in its entirety for all purposes. This application is a nonprovisional of and claims priority to U.S. Patent application No: 60/838,452, filed August 17, 2006, entitled "Polysaccharide Compositions and Methods of Administering, Producing, and Formulating Polysaccharide Compositions", which is hereby incorporated in its entirety for all purposes. This application is a nonprovisional of and claims priority to U.S. Patent application No: 60/816,967, filed June 28, 2006, entitled "Zeaxanthin Production Methods and Novel Compositions Containing Zeaxanthin", which is hereby incorporated in its entirety for all purposes. This application is a nonprovisional of and claims priority to U.S. Patent application No: 60/872,072, filed November 30, 2006, entitled "Polysaccharide Compositions and Methods of Administering, Producing, and Formulating Polysaccharide Compositions", which is hereby incorporated in its entirety for all purposes. U.S. Patent application No: , filed January 19,
2007, entitled "Microalgae-derived Compositions for Improving the Health and Appearance of Skin", is hereby incorporated in its entirety for all purposes. AU other references cited are incorporated in their entirety for all purposes.
Definitions: The following definitions are intended to convey the intended meaning of terms used throughout the specification and claims, however they are not limiting in the sense that minor or trivial differences fall within their scope.
"Active in microalgae" means a nucleic acid that is functional in microalgae. For example, a promoter that has been used to drive an antibiotic resistance gene to impart antibiotic resistance to a transgenic microalgae is active in microalgae. Nonlimiting examples of promoters active in microalgae are promoters endogenous to certain algae species and promoters found in plant viruses.
"Antiviral lubricant" means a molecule that possesses both antiviral activity and lubricant activity.
"ARA" means Arachidonic acid.
"Associates with" means, within the context of a polysaccharide binding fusion protein, one molecule binding to another molecule. Affinity and selectivity of binding can vary when a polysaccharide and a polysaccharide binding protein are in association with each other. "Axenic" means a culture of an organism that is free frqm contamination by other living organisms.
"Bioreactor" means an enclosure or partial enclosure in which cells are cultured in suspension.
"Carbohydrate modifying enzyme" means an enzyme that utilizes a carbohydrate as a substrate and structurally modifies the carbohydrate.
"Carbohydrate transporter" means a polypeptide that resides in a lipid bilayer and facilitates the transport of carbohydrates across the lipid bilayer.
"Carrier suitable for human consumption" refers to compounds and materials suitable for human ingestion or otherwise physiologically compatible with oral administration to humans. Usually, such carriers are of plant or animal origin. Although such carriers sometimes contain residual amounts of solvents and buffers used in the processing of the polysaccharides and other compositions of the invention, they do not consist exclusively of such solvents or buffers, and usually have less than 50% and preferably less than 10% w/w of such solvents or buffers.
"Carrier suitable for topical administration" means a compound that may be administered, together with one or more compounds of the present invention, and which does not destroy the activity thereof and is nontoxic when administered in concentrations and amounts sufficient to deliver the compound to the skin or a mucosal tissue.
"Cell material" means any matter obtained from cells. Cell material can be intact cells, disrupted cells, a purified componenet of cells, and extract of cells, and other materials.
"Combination Product" means a product that comprises at least two distinct compositions intended for human administration through distinct routes, such as a topical route and an oral route. In some embodiments the same active agent is contained in both the topical and oral components of the combination product.
As used herein, "comprise" or "comprising" or "include" or "including" or variants thereof are used in the "open" sense such that the terms are inclusive and permit the presence of additional elements. The terms specify the presence of the stated features- steps, or components as recited without precluding the presence or addition of one or more features, steps, or components.
"Conditions favorable to cell division" means conditions in which cells divide at least once every 72 hours. "Derived from" means, within the context of a microalgae strain, a strain that has been generated through passaging or mutagenesis from a starting strain. For example, Porphyridium strain UTEX 637 can be subjected to chemical mutagenesis to generate new colonies. Each new colony is a strain deruived from Porphyridium strain UTEX 637.
"DHA" means Docosahexaenoic acid.
"Endopolysaccharide" means a polysaccharide that is retained intracellularly.
"EPA" means eicosapentaenoic acid.
"Exogenous gene" means agene transformed into a wild-type organism. The gene can be heterologous from a different species, or homologous from the same species, in which case the gene occupies a different location in the genome of the organism than the endogenous gene.
"Exogenously provided" describes a molecule provided to the culture media of a cell culture.
"Exopolysaccharide" means a polysaccharide that is secreted from a cell into the extracellular environment.
"Filtrate" means the portion of a tangential flow filtration sample that has passed through the filter.
"Fixed carbon source" means molecule(s) containing carbon that are present at ambient temperature and pressure in solid or liquid form.
"Glycopolymer" means a biologically produced molecule comprising at least two monosaccharides. Examples of glycopolymers include glycosylated proteins, polysaccharides, oligosaccharides, and disaccharides.
A coding sequence is described as "heterologous" when it is physically connected to a nucleic acid other than that which it is naturally connected to in a wild-type genome. A coding sequence not found in a wild-type genome is necessarily heterologous to a promoter that it is in operable linkage with.
"Homogenate" means cell biomass that has been disrupted. A homogenate is not necessarily completely homogeneous, and as used herein can refer to a composition of cells that have been disrupted to the point where a majority of the cells in the preparation have been ruptured. A homogenate may contain chunks of cell wall, intact or partially intact organelles such as chloroplasts and mitochondria, and other substructures of the cell.
"Mammal" as used herein includes animal subjects, such as primates, as well has human patients. "Microalgae" means a single-celled organism that is capable of performing photosynthesis. Microalgae include obligate photoautotrophs, which cannot metabolize a fixed carbon source as energy, as well as heterotrophs, which can live solely off of light, solely off of a fixed carbon source, or a combination of the two.
"Naturally produced" describes a compound that is produced by a wild-type organism.
"Pharmaceutically acceptable carrier or adjuvant" refers to a carrier or adjuvant that may be administered to a patient, together with one or more compounds of the present invention, and which does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the compound. "Photobioreactor" means a waterproof container, at least part of which is at least partially transparent, allowing light to pass through, in which one or more microalgae cells are cultured. Photobioreactors may be sealed, as in the instance of a polyethylene bag, or may be open to the environment, as in the instance of a pond.
"Polysaccharide material" is a composition that contains more than one species of polysaccharide, and optionally contaminants such as proteins, lipids, and nucleic acids, such as, for example, a microalgal cell homogenate.
"Polysaccharide" means a compound or preparation containing one or more molecules that contain at least two saccharide molecules covalently linked. A "polysaccharide", "endopolysaccharide" or "exopolysaccharide" can be a preparation of polymer molecules that have similar or identical repeating units but different molecular weights within the population.
"Port", in the context of a photobioreactor, means an opening in the photobioreactor that allows influx or efflux of materials such as gases, liquids, and cells. Ports are usually connected to tubing leading to and/or from the photobioreactor.
"Red microalgae" means unicellular algae that is of the list of classes comprising Bangiophyceae, Florideophyceae, Goniotrichales, or is otherwise a member of the Rhodophyta.
"Retentate" means the portion of a tangential flow filtration sample that has not passed through the filter.
"Selectively binds to" refers to a binding reaction which is determinative of the presence of a molecule in the presence of a heterogeneous population of other molecules. Thus, under designated conditions, a specified ligand binds preferentially to a particular molecule and does not bind in a significant amount to other proteins present in the sample. A molecule such as antibody that specifically binds to a protein often has an association constant of at least 106M'1 or 107 M"1, preferably 108 M"1 to 109 M'1, and more preferably, about 1010 M"1 to 101 ' M"1 or higher. A variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein. For example, solid- phase ELISA immunoassays are routinely used to select monoclonal antibodies specifically immunoreactive with a protein. See, e.g., Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, New York, for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity.
"Small molecule" means a molecule having a molecular weight of less than 2000 daltons, in some instances less than 1000 daltons, and in still other instances less than 500 daltons or less. Such molecules include, for example, heterocyclic compounds, carbocyclic compounds, sterols, amino acids, lipids, carotenoids and polyunsaturated fatty acids.
A molecule is "solvent available" when the molecule is isolated to the point at which it can be dissolved in a solvent, or sufficiently dispersed in suspension in the solvent such that it can be detected in the solution or suspension. For example, a polysaccharide is "solvent available" when it is sufficiently isolated from other materials, such as those with which it is naturally associated, such that the polysaccharide can be dissolved or suspended in an aqueous buffer and detected in solution using a dimethylmethylene blue (DMMB) or phenohsulfuric acid assay. In the case of a high molecular weight polysaccharide containing hundreds or thousands of monosaccharides, part of the polysaccharide can be "solvent available" when it is on the outermost layer of a cell wall while other parts of the same polysaccharide molecule are not "solvent available" because they are buried within the cell wall. For example, in a culture of microalgae in which polysaccharide is present in the cell wall, there is little "solvent available" polysaccharide since most of the cell wall polysaccharide is sequestered within the cell wall and not available to solvent. However, when the cells are disrupted, e.g., by sonication, the amount of "solvent available" polysaccharide increases. The amount of "solvent accessible" polysaccharide before and after homogenization can be compared by taking two aliquots of equal volume of cells from the same culture, homogenizing one aliquot, and comparing the level of polysaccharide in solvent from the two aliquots using a DMMB assay. The amount of solvent accessible polysaccharide in a homogenate of cells can also be compared with that present in a quantity of cells of the same type in a different culture needed to generate the same amount of homogenate. "Substantially free of protein" means compositions that are preferably of high purity and are substantially free of potentially harmful contaminants, including proteins (e.g., at least National Food (NF) grade, generally at least analytical grade, and more typically at least pharmaceutical grade). Compositions are at least 80, at least 90, at least 99 or at least 99.9% w/w pure of undesired contaminants such as proteins are substantially free of protein. To the extent that a given compound must be synthesized prior to use, the resulting product is typically substantially free of any potentially toxic agents, particularly any endotoxins, which may be present during the synthesis or purification process. Compositions are usually made under GMP conditions. Compositions for parenteral administration are usually sterile and substantially isotonic.
I General
Polysaccharides form a heterogeneous group of polymers of different length and composition. They are constructed from monosaccharide residues that are linked by glycosidic bonds. Glycosidic linkages may be located between the Ci (or C2) of one sugar residue and the C2, C3, C4, C5 or Ce of the second residue. A branched sugar results if more than two types of linkage are present in single monosaccharide molecule.
Monosaccharides are simple sugars with multiple hydroxyl groups. Based on the number of carbons (e.g., 3, 4, 5, or 6) a monosaccharide is a triose, tetrose, pentose, or hexose. Pentoses and hexoses can cyclize, as the aldehyde or keto group reacts with a hydroxyl on one of the distal carbons. Examples of monosaccharides are galactose, glucose, and rhamnose.
Polysaccharides are molecules comprising a plurality of monosaccharides covalently linked to each other through glycosidic bonds. Polysaccharides consisting of a relatively small number of monosaccharide units, such as 10 or less, are sometimes referred to as oligosaccharides. The end of the polysaccharide with an anomeric carbon (Ci) that is not involved in a glycosidic bond is called the reducing end. A polysaccharide may consist of one monosaccharide type, known as a homopolymer, or two or more types of monosaccharides, known as a heteropolymer. Examples of homopolysaccharides are cellulose, amylose, inulin, chitin, chitosan, amylopectin, glycogen, and pectin. Amylose is a glucose polymer with α(l ->4) glycosidic linkages. Amylopectin is a glucose polymer with α(l ->4) linkages and branches formed by α(l ->6) linkages. Examples of heteropolysaccharides are glucomannan, galactoglucomannan, xyloglucan, 4-O- methylglucuronoxylan, arabinoxylan, and 4-O-Methylglucuronoarabinoxylan. Polysaccharides can be structurally modified both enzymatically and chemically. Examples of modifications include sulfation, phosphorylation, methylation, O-acetylation, fatty acylation, amino N-acetylation, N-sulfation, branching, and carboxyl lactonization.
Glycosaminogl yeans are polysaccharides of repeating disaccharides. Within the disaccharides, the sugars tend to be modified, with acidic groups, amino groups, sulfated hydroxyl and amino groups. Glycosaminoglycans tend to be negatively charged, because of the prevalence of acidic groups. Examples of glycosaminoglycans are heparin, chondroitin, and hyaluronic acid.
Polysaccharides are produced in eukaryotes mainly in the endoplasmic reticulum (ER) and Golgi apparatus. Polysaccharide biosynthesis enzymes are usually retained in the ER, and amino acid motifs imparting ER retention have been identified (Gene. 2000 Dec 31;261(2):321-7). Polysaccharides are also produced by some prokaryotes, such as lactic acid bacteria.
Polysaccharides that are secreted from cells are known as exopolysaccharides. Many types of cell walls, in plants, algae, and bacteria, are composed of polysaccharides. The cell walls are formed through secretion of polysaccharides. Some species, including algae and bacteria, secrete polysaccharides that are released from the cells. In other words, these molecules are not held in association with the cells as are cell wall polysaccharides. Instead, these molecules are released from the cells. For example, cultures of some species of microalgae secrete exopolysaccharides that are suspended in the culture media.
II Methods of Producing Polysaccharides
A. Cell Culture Methods: Microalgae
Polysaccharides can be produced by culturing microalgae. Examples of microalgae that can be cultured to produce polysaccharides are shown in Table 1. Also listed are references that enable the skilled artisan to culture the microalgae species under conditions sufficient for polysaccharide production. Also listed are strain numbers from various publicly available algae collections, as well as strains published in journals that require public dissemination of reagents as a prerequisite for publication.
Table 1
Microalgae are preferably cultured in liquid media for polysaccharide production. Culture condition parameters can be manipulated to optimize total polysaccharide production as well as to alter the structure of polysaccharides produced by microalgae.
Microalgal culture media usually contains components such as a fixed nitrogen source, trace elements, a buffer for pH maintenance, and phosphate. Other components can include a fixed carbon source such as acetate or glucose, and salts such as sodium chloride, particularly for seawater microalgae. Examples of trace elements include zinc, boron, cobalt, copper, manganese, and molybdenum in, for example, the respective forms of ZnCl2, H3BO3, CoCl2-6H2O, CuCl2-2H2O, MnCl2-4H2O and (NH4)6Mo7θ24-4H2O.
Some microalgae species can grow by utilizing a fixed carbon source such as glucose or acetate. Such microalgae can be cultured in bioreactors that do not allow light to enter. Alternatively, such microalgae can also be cultured in photobioreactors that contain the fixed carbon source and allow light to strike the cells. Such growth is known as heterotrophic growth. Any strain of microalgae, including those listed in Table 1, can be cultured in the presence of any one or more fixed carbon source including those listed in Tables 2 and 3.
Table 2
2,3-Butanediol 2-Aminoethanol
2'-Deoxy Adenosine
3 -Methyl Glucose
Acetic Acid
Adenosine
Adenosine-5 '-Monophosphate
Adonitol
Amygdalin
Arbutin
Bromosuccinic Acid
Cis-Aconitic Acid
Citric Acid
D,L-Carnitine
D,L-Lactic Acid
D,L-α-Glycerol Phosphate
D- Alanine
D-Arabitol
D-Cellobiose
Dextrin
D-Fructose
D-Fructose-6-Phosphate
D-Galactonic Acid Lactone
D -Galactose
D-Galacturonic Acid
D-Gluconic Acid
D-Glucosaminic Acid
D-Glucose-6-Phosphate
D-Glucuronic Acid
D-Lactic Acid Methyl Ester
D-L-α-Glycerol Phosphate
D-Malic Acid
D-Mannitol
D-Mannose
D-Melezitose
D-Melibiose
D-Psicose
D-Raffinose
D-Ribose
D-Saccharic Acid
D-Seririe
D-Sorbitol
D-Tagatose
D-Trehalose
D-Xylose
Formic Acid
Gentiobiose
Glucuronamide
Glycerol
Glycogen
Glycyl-LAspartic Acid Glycyl-LGlutamic Acid
Hydroxy-LProline i-Erythritol
Inosine
Inulin
Itaconic Acid
Lactamide
Lactulose
L-Alaninamide
L- Alanine
L-Alanylglycine
L-Al anyl-Glycine
L-Arabinose
L-Asparagine
L-Aspartic Acid
L-Fucose
L-Glutamic Acid
L-Histidine
L-Lactic Acid
L-Leucine
L-Malic Acid
L-Ornithine
LPhenylalanine
L-Proline
L-Pyroglutamic Acid
L-Rharnnose
L-Serine
L-Threonine
Malonic Acid
Maltose
Maltotriose
Mannan m-Inositol
N-Acetyl-DGalactosamine
N-Acetyl-DGlucosamine
N- Acetyl- LGlutamic Acid
N-Acetyl-β-DMannosamine
Palatinose
Phenyethylamine p-Hydroxy-Phenylacetic Acid
Propionic Acid
Putrescine
Pyruvic Acid
Pyruvic Acid Methyl Ester
Quinic Acid
Salicin
Sebacic Acid
Sedoheptulosan
Stachyose
Succinamic Acid Succinic Acid
Succinic Acid Mono-Methyl-Ester
Sucrose
Thymidine
Thymidine-5'-Monophosphate
Turanose
Tween 40
Tween 80
Uridine
Uridine-5'-Monophosphate
Urocanic Acid
Water
Xylitol α-Cyclodextrin α-D-Glucose α-D-Glucose- 1 -Phosphate α-D-Lactose α-Hydroxybutyric Acid α-Keto Butyric Acid α-KLeto Glutaric Acid α-Keto Valeric Acid α-Ketoglutaric Acid α-Ketovaleric Acid α-Methyl-DGalactoside α-Methyl-DGlucoside α-Methyl-DMannoside β-Cyclodextrin β-Hydroxybutyric Acid β-Methyl-DGalactoside β-Methyl-D-Glucoside γ- Amino Butyric Acid γ-Hydroxybutyric Acid
Table 3
(2-amino-3 ^-dihydroxy-S-hydroxymethyl- 1 -cyclohexyl)glucopyranoside
(S^-disinapoy^fi-uctoftiranosyl-Cό-sinapoy^glucopyranoside
(3-sinapoyl)fructofliranosyl-(6-sinapoyl)glucopyrariOside
1 reference
1 , 10-di-O-(2-acetamido-2-deoxyglucopyranosyl)-2-azi- 1 , 10-decanediol
1 ,3-mannosylinannose
1 ,6-anhydrolactose
1,6-anhydrolactose hexaacetate
1 ,6-dichIorosucrose
1 -chlσrosucrose
1 -desoxy-1 -glycinomaltose l-O-alpha^-acetamido^-deoxygalactopyranosyl-inositol
1 -O-methyl-di-N-tri fluoroacetyl-beta-chitobioside l-propyl-4-O-beta galactopyranosyl -alpha galactopyranoside
2-(acetylamino)-4-O-(2-(acetylamino)-2-deoxy-4-O-sulfogalactopyranosyl)-2-deoxyglucose 2-(trimethylsilyl)ethyl lactoside
2,ll,3',4',6'~penta-O-acetylsucrose
2,2'-0-(2,2'-diacetamido-2,3;2',3'-tetradeoxy-656'-di-0-(2-tetradecylhexadecanoyl)- alpha,alpha'-trehalose-3,3'-diyl)bis(N-lactoyl-alanyl-isogliιtaniine)
2,3,6,2',3',4I,6l-hepta-O-acetylcellobiose
2,3'-anhydrosucrose
2,3-di-O-phytanyI- 1 -O-(mannopyranosyl-(2-sulfate)-( 1 -2)-glucoρyranosyl)-sn-glycerol
2,3-epoxypropyl O-galactopyranosyl(l-6)galactopyranoside
2,3-isoprolylideneerthrofuranosyl 2,3-O-isopropylideneerythrofuranoside
2',4'-dinitrophenyl 2-deoxy-2-fluoro-beta-xylobioside
2,5-anhydromannitol iduronate
2,6-sialyllactose
2-acetamido-2,4-dideoxy-4-fluoro-3-0-galactoρyranosylglucopyranose
2-acetamido-2-deoxy-3-O-(gluco-4-enepyranosyluronic acid)glucose
2-acetamido-2-deoxy-3-O-rhamnopyranosylglucose
2-acetamido-2-deoxy-6-0-beta galactopyranosylgalactopyranose
2-acetamido-2-deoxyglucosylgalactitol
2-acetamido-3-0-(3-acetamido-3,6-dideoxy-beta-glucopyranosyl)-2-deoxy-galactopyranose
2-amino-6-0-(2-amino-2-deoxy-glucopyranosyl)-2-deoxyglucose
2-azido-2-deoxymannopyranosyl-(l,4)-rhamnopyranose
2-deoxy-6-0-(2,3-dideoxy-4,6-0-isopropylidene-2,3-(N-tosylepimino)mannopyranosyl)-
4,5-O-isopropylidene- 1 ,3 -di-N-tosylstreptamine
2-deoxymaltose
2-iodobenzyl-l -thiocellobioside
2-N-(4-benzoyl)benzoyl- 1 ,3-bis(mannos-4-yloxy)-2-propylamine
2-nitrophenyl-2-acetamido-2-deoxy-6-O-beta galactopyranosyl-alpha galactopyranoside
2-O-(glucopyranosyluronic acid)xylose
2-O-glucopyranosylribitol- 1 -phosphate
2-0-glucopyranosylribitol-4'-phosphate
2-0-rhamnopyraπosyl-rhaπxnopyraπosyl-3-hydroxyldecanoyl-3-hydroxydecanoate
2-O-talopyranosylmannopyranoside
2-thiokojibiose
2-tbiosophorose
3,3'-neotxehalosadiamine
3,6,3',6'-dianhydro(galactopyranosylgalactopyranoside)
3,6-di-O-methyl-beta-glucopyranosyl-(l-4)-2,3-di-O-meth.yl-alpha-rhamnopyranose
3 -amino-3 -deoxyaltropyranosyl-3 -amino-3 -deoxyaltropyranosi de
3 -deoxy-3 -fluorosucrose
3-deoxy-5-O-rhamnopyranosyl-2-octuloρyranosonate
3-deoxyoctulosonic acid-(alpha-2-4)-3-deoxyoctulosonic acid
3-deoxysucrose
3-ketolactose
3-ketosucrose
3-ketotrehalose
3-methyllactose
3-0-(2-acetamido-6-0-(N-acetylneuraminyl)-2-deoxygalactosyl)serine
3-O-(glucopyranosyluronic acid)galactoρyranose
3 -O-beta-glucuronosylgalactose
S-O-fiicopyranosyl^-acetamido^-deoxyglucopyranose
3'-O-galactopyranosyl- 1 -4-O-galactopyranosylcytarabine 3-O-galactosylarabinose
3 -O-talopyranosylmannopyranoside
3 -trehalosamine
4-(trifluoroacetamido)phenyl-2-acetamido-2-deoxy-4-O-beta-mannopyranosyl-beta- glucopyranoside
4,4',6,6'-tetrachloro-454',6,6l-tetradeoxygalactotrehalose
4,6,4',6'-dianhydro(galactopyranosylgalactopyranoside)
4,6-dideoxysucrose
4,6-O-(l -ethoxy-2-propenylidene)sucrose hexaacetate
4-chloro-4-deoxy-alpha-gaIactoρyranosyl 3 ,4-anhydro- 1 ,6-dichloro- 1 ,6-dideoxy-beta-lyxo- hexulofuranoside
4-glucopyranosylmannose
4-methylumbelliferylcellobioside
4-nitrophenyl 2-fucopyranosyl-fucopyranoside
4-nitrophenyl 2-O-alpha-D-galactopyranosyl-alρha-D-mannopyranoside
4-nitrophenyl 2-O-alpha-D-glucopyranosyl-alpna-D-mannopyranoside
4-nitrophenyl 2-O-alpha-D-mannoρyτanosyl-alpha-D- mannopyranoside
4-nitrophenyl 6-O-alpha-D-mannopyranosyl-alρha-D-mannopyranoside
4-nitrophenyl-2-acetamido-2-deoxy-6-O-beta-D- galactopyranosyl-beta-D-glucopyranoside
4-O-(2-acetarnido-2-deoxy-beta-glucopyranosyl)ribitol
4-O-(2-amino-2-deoxy-alpha-glucopyranosyl)-3-deoxy-manno-2-octulosonic acid
4-O-(glucoρyranosyluronic acid)xylose
4-O-acetyl-alpha-N-acetylneuraminyl-(2-3)-lactose
4-O-alpha-D-galactopyranosyl-D-galactose
4-O-galactopyranosyl-3,6-anhydrogalactose dimethylacetal
4-O-galactopyranosylxylose
4-O-mannopyranosyl-2-acetamido-2-deoxyglucose
4-thioxylobiose
4-trehalosamine
4-trifluoroacetamidophenyl 2-acetamido-4-O-(2-acetamido-2-deoxyglucopyranosyl)-2- deoxymannopyranosiduronic acid
5-bromoindoxyl-beta-cellobioside
5'-0-(fructofuranosyl-2- 1 -fructofuranosyl)pyridoxine
S-O-beta-galactofuranosyl-galactofuranose
6 beta-galactinol
6(2)-thiopanose δjό'-di-O-coiynomycoloyl-alpha-mannopyranosyl-alpha-mannopyranoside όjό-di-O-maltosyl-beta-cyclodextrin δjό'-di-O-mycoloyl-alpha-mannopyranosyl-alpha-marmopyranoside
6-chloro-6-deoxysucrose
6-deoxy-6-flιxorosucrose ό-deoxy-alpha-gluco-pyranosiduronic acid ό-deoxy-gluco-heptopyranosyl ό-deoxy-gluco-heptopyranoside
6-deoxysucrose
6-0-decanoyl-3,4-di-0-isobutyrylsucrose ό-O-galactopyranosyl^-acetamido^-deoxygalactose
6-O-galactoρyranosyl galactose
6-O-heptopyranosylglucopyranose
6-thiosucrose
7-0-(2-amino-2-deoxyglucoρyranosyl)heptose δ-methoxycarbonyloctyl-S-O-glucopyranosyl-mannopyranoside
8-0-(4-amino-4-deoxyarabinopyranosyl)-3 -deoxyoctulosonic acid allolactose allosucrose allyl 6-O-(3-deoxyoct-2-ulopyranosylonic acid)-(l -6)-2-deoxy-2-(3- hydroxytetradecanamido)glucopyranoside 4-phosphate alpha-(2-9)-disialic acid alpha,alpha-trehalose 6,6'-diphosphate alpha-glucopyranosyl alpha-xylopyranoside alpha-maltosyl fluoride aprosulate benzyl 2-acetamido-2-deoxy-3-0-(2-0-methyl-beta-galactosyl)-beta-glucopyτanoside benzyl 2-acetamido-2-deoxy-3-0-beta fucopyranosyl-alpha-galactopyranoside benzyl 2-acetamido-6-0-(2-acetamido-2,4-dideoxy-4-fluoroglucopyranosyl)-2- deoxygalactopyranoside benzyl gentiobioside beta-D-galactosyl(l-3)-4-nitrophenyl-N-acetyl-alρha-D-galactosamine beta-methylmelibiose calcium sucrose phosphate camiglibose cellobial cellobionic acid cellobionolactone
Cellobiose cellobiose octaacetate cellobiosyl bromide heptaacetate
Celsior chitobiose chondrosine
Cristolax deuterated methyl beta-mannobioside dextrin maltose
D-glucopyranose, O-D-glucopyranosyl
Dietary Sucrose difructose anhydride I difructose anhydride III difructose anhydride IV digalacturonic acid
DT 5461 ediol epilactose epsilon-N-l-Cl-deoxylactulosyl^ysine feruloyl arabinobiose floridoside fructofuranosyl-(2-6)-glucopyranoside
FZ 560 galactosyl -( 1 -3)galactose garamine gentiobiose geranyl 6-O-alpha-L-arabinopyranosyl-beta-D-glucopyranoside geranyl 6-O-xyloρyraiiosyl-glucopyranoside glucosaminyl-1 ,6-inositol-l,2-cyclic monophosphate glucosyl (1-4) N-acetylglucosamine glucuronosyl(l -4)-rhamnose heptosyl-2-keto-3 -deoxyoctonate inulobiose
Isomaltose isomaltulose isoprimeverose kojibiose lactobionic acid lacto-N-biose II
Lactose lactosylurea
Lactulose laminaribiose lepidimoide leucrose levanbiose hicidin 3-O-beta-primveroside
LW 10121
LW 10125
LW 10244 maltal maltitol
Maltose maltose hexastearate maltose-maleimide maltosylnitromethane heptaacetate maltosyltriethoxycholesterol maltotetraose
Malun 25 mannosucrose maπnosyl-(l -4)-N-acetylglucosaminyl-( 1 -N)-urea maimosyl(2)-N-acetyl(2)-glucose melibionic acid
Melibiose melibiouronic acid methyl 2-acetamido-2-deoxy-3 -O-(alpha-idopyranosyluronic acid)-4-O-sulfo-beta- galactopyranoside methyl 2-acetamido-2-deoxy-3-0-(beta-glucopyranosyluronic acid)-4-0-suIfo-beta- galactopyranoside methyl 2-acetamido-2-deoxy-3-O-glucoρyranosyluronoylglucopyranoside methyl 2-O-alρha-rhamnopyranosyl-beta-galactopyranoside methyl 2-O-beta-rhamnopyranosyl-beta-galactoρyranoside methyl 2-O-fucopyranosylracopyranoside 3 sulfate methyl 2-O-mannopyranosylmannopyranoside methyl 2-O-mannopyranosyl-rhamnopyranoside methyl 3-O-(2-acetamido-2-deoxy-6-thioglucopyranosyl)galactoρyranoside methyl 3-O-galactopyranosylmannopyranoside methyl 3 -O-mannopyranosylmannopyranoside methyl 3-O-mannopyranosyltalopyranoside methyl 3-O-talopyranosyltalopyranoside methyl 4-0-(6-deoxy-manno-heptopyranosyl)galactopyranoside methyl 6-0-acetyl-3-0-benzoyl-4-0-(2,3,4,6-tetra-0-benzoylgalactopyranosyl)-2-deoxy-2- phthalimidoglucopyranoside - methyl 6-O-mannopyranosylmannopyranoside methyl beta-xylobioside methyl fijcopyranosyl(l -4)-2-acetamido-2-deoxyglucopyranoside methyl laminarabioside methyl O-(3 -deoxy-3-fluorogalactopyranosyl)(l -6)galactopyranoside methyl-2-acetamido-2-deoxyglucopyranosyl- 1 -4-glucopyranosiduronic acid methyl-2-O-fucopyranosylfucopyranoside 4-sulfate
MK 458
N(l)-2-carboxy-4,6-dinitrophenyl-N(6)-lactobionoyl-l,6-hexanediamine
N-(2 ,4-dinitro-5-fluoropheπyl)- 1 ,2-bis(mannos-4'-yloxy)propyl-2-amine
N-(2'-mercaρtoethyl)lactamine
N-(2 -nitro-4-azoρhenyl)- 1 ,3 -bi s(manno s-4'-yloxy)propyl-2-amine
N-(4-azidosalicylamide)- 1 ,2-bis(mannos-4'-yloxy)propyl-2-amine
N,N-diacetylchitobiose
N-acetylchondrosine
N-acetyldermosine
N-acetylgalactosaminyl-(l-4)-galactose
N-acetylgalactosaminyl-(l-4)-glucose
N-acetylgalactosaminyl- 1 -4-N-acetyIgIucosamine
N-acetylgalactosaminyl- 1 -4-N-acetylglucosamine
N-acetylgalactosaminyl-alpha(l-3)galactose
N-acetylglυcosamine-N-acetylmuramyl-alanyl-isoglutaminyl-alanyl-glycerol dipalmitoyl
N-acetylglucosaminyl beta(l -6)N- acetylgalactosamine
N-acetylglucosaminyl- 1 -2-mannopyranose
N-acetylhyalobiuronic acid
N-acetylneuraminoyllactose
N-acetylneviraminoyllactose sulfate ester
N-acetylneuraminyl-(2-3)-galactose
N-acetylneταraminyl-(2-6)-galactose
N-benzyl-4-0-(beta-galactopyranosyl)glucamine-N-carbodithioate neoagarobiose
N-formylkansosaminyl-(l-3)-2-O-methylrhamnopyranose
0-((Nalpha)-acetylglucosamine 6-sulfate)-(l-3)-idonic acid
O-(4-O-feruloyl-alpha-xyiopyranosyl)-(l-6)-glucopyranose
O-(alpha-idopyranosyluronic acid)-(l-3)-2,5-anhydroalditol-4-sulfate
O-(glucuronic acid 2-sulfate)-(l~3)-0-(2,5)-andydrotalitol 6-sulfate .
O-(glucuronic acid 2-sulfate)-(l~4)-0-(2,5)-anhydromannitol 6-sulfate
0-alpha-glucopyranosyluronate-(l-2)-galactose
O-beta-galactopyranosyl-(l-4)-O-beta-xylopyranosyl-(l-0)-serine octyl maltopyranoside
O-demethylpauloinycin A
O-demethylpaulomycin B
O-methyl-di-N-acetyl beta-chitobioside
Palatimt paldimycin paulomenol A paulomenol B paulomycin A paulomycin A2 paulomycin B paulomycin C paulomycin D paulomycin E paulomycin F phenyl 2-acetamido-2-deoxy-3-0-beta-D-galactopyranosyl-alpha-D-galactoρyranoside phenyl 0-(2,3,4s6-tetra-0-acetylgalactopyranosyl)-(l-3)-4,6-di-0-acetyl-2-deoxy-2- phthalimido- 1 -thioglucopyranoside poly-N-4-vinylbenzyllactonamide pseudo-cellobiose pseudo-maltose rhamnopyranosyl-(l-2)-rhamnopyranoside-(l-methyl ether) rhoifolin ruberythric acid
S-3105 senfolomycin A senfolomycin B solabiose
SS 554 streptobiosamine
Sucralfate
Sucrose sucrose acetate isobutyrate sucrose caproate sucrose distearate sucrose monolaurate sucrose monopalmitate sucrose monostearate sucrose myristate sucrose octaacetate sucrose octabenzoic acid sucrose octaisobutyrate sucrose octasulfate sucrose polyester sucrose sulfate swertiamacroside
T-1266 tangshenoside I tetrahydro-2-((tetrahydro-2-furanyl)oxy)-2H-pyran thionigerose
Trehalose trehalose 2-sulfate trehalose 6,6'-dipalmitate trehalose-6-phosphate trehalulose trehazolin trichlorosucrose tunicamine turanose
U 77802
U 77803 xylobiose xylose-glucose xylosucrose
Microalgae contain photosynthetic machinery capable of metabolizing photons, and transferring energy harvested from photons into fixed chemical energy sources such as monosaccharide. Glucose is a common monosaccharide produced by microalgae by metabolizing light energy and fixing carbon from carbon dioxide. Some microalgae can also grow in the absence of light on a fixed carbon source that is exogenously provided (for example see Plant Physiol. 2005 Feb;137(2):460-74). In addition to being a source of chemical energy, monosaccharides such as glucose are also substrate for production of polysaccharides (see Example 14). The invention provides methods of producing polysaccharides with novel monosaccharide compositions. For example, microalgae is cultured in the presence of culture media that contains exogenously provided monosaccharide, such as glucose. The monosaccharide is taken up by the cell by either active or passive transport and incorporated into polysaccharide molecules produced by the cell. This novel method of polysaccharide composition manipulation can be performed with, for example, any microalgae listed in Table 1 using any monosaccharide or disaccharide listed in Tables 2 or 3.
In some embodiments, the fixed carbon source is one or more selected from glucose, galactose, xylose, mannose, rhamnose, N-acetylglucosamine, glycerol, floridoside, and glucuronic acid. The methods may be practiced cell growth in the presence of at least about 5.0 μM, at least about 10 μM, at least about 15.0 μM, at least about 20.0 μM, at least about 25.0 μM, at least about 30.0 μM, at least about 35.0 μM, at least about 40.0 μM, at least about 45.0 μM, at least about 50.0 μM, at least about 55.0 μM, at least about 60.0 μM, at least about 75.0 μM, at least about 80.0 μM, at least about 85.0 μM, at least about 90.0 μM, at least about 95.0 μM, at least about 100.0 μM, or at least about 150.0 μM, of one or more exogenously provided fixed carbon sources selected from Tables 2 and 3.
In some embodiments using cells of the genus Porphyridium, the methods include the use of approximately 0.5-0.75% glycerol as a fixed carbon source when the cells are cultured in the presence of light. Alternatively, a range of glycerol, from approximately 4.0% to approximately 9.0% may be used when the Porphyridium cells are cultured in the dark, more preferably from 5.0% to 8.0%, and more preferably 7.0%.
After culturing the microalgae in the presence of the exogenously provided carbon source, the monosaccharide composition of the polysaccharide can be analyzed as described in Example 5. Microalgae can be transformed with genes encoding carbohydrate transporters to facilitate the uptake of exogenously provided carbohydrates such SEQ ID NOs: 20, 22, 24, 26, 27, 29-39 and 46-48.
Microalgae culture media can contain a fixed nitrogen source such as KNO3. Alternatively, microalgae are placed in culture conditions that do not include a fixed nitrogen source. For example, Porphyridium sp. cells are cultured for a first period of time in the presence of a fixed nitrogen source, and then the cells are cultured in the absence of a fixed nitrogen source (see for example Adda M., Biomass 10:131-140. (1986); Sudo H., et al., Current Microbiology Vol. 30 (1995), pp. 219-222; Marinho-Soriano E., Bioresour Technol. 2005 Feb;96(3):379-82; Bioresour. Technol. 42:141-147 (1992)).
Other culture parameters can also be manipulated, such as the pH of the culture media, the identity and concentration of trace elements such as those listed in Table 3, and other media constituents. One non-limiting example is the inclusion of at least one source of sulfate in the culture media to increase the level of sulfation in the polysaccharides produced. In some embodiments, a polysaccharide preparation method is practiced with culture media containing more than about 100, more than about 150, more than about 200, more than about 250, more than about 300, more than about 350, more than about 400, more than about 450, more than about 500, more than about 550, more than about 600, more than about 650, more than about 700, or more than about 750, more than about 800, more than about 850, more than about 900, more than about 950, or more than about 1 or 2 M sulfate (or total SO4 2"). Increasing the sulfation has been demonstrated to increase the antioxidant apacity of the polysaccharide (see example 23 and Spitz et al. (J. Appl. Phycology (2005) 17:215-222). Without being bound by theory, and offered to improved the understanding of certain aspects of the disclosed invention, it is possible that an increased level of sulfation may increase the anti-cholesterol characteristics of the homogenized cell material or polysaccharide preparation disclosed herein. The correlation between higher amounts of sulfation and antioxidant activity demonstrated herein was unexpected given the weak antioxidant activity of carrageenan, which contains as much as 40% sulfate. It is believed that rαicroalgae of the genus Porphyridiu'm have not been grown or propagated under conditions with sulfate concentrations of 100 mM to 2 M. Thus the invention includes the surprising discovery that microalgae are capable of growth under such conditions. Additionally, the invention is based in part on the surprising discovery that growth under higher sulfate concentrations can produce polysaccharides with higher levels of sulfation. This allows for the production of cells (and so biomass) containing highly sulfated polysaccharides that may be used in the form of purified polysaccharides, a homogenate of cells (biomass), intact cells (biomass) per se, or a combination thereof.
The discovery that Porphyridiiim can survive above 10OmM sulfate was surprising for a number of reasons. First, it is known that sulfate can alter the rate of uptake of toxic metal ions in algae, such as chromium, and that increasing metal accumulation can lead to toxicity (see for example Kaszycki et al, Plant, Cell & Environment, 28(2): p.260, 2005). Second, it is also known that sulfate can inhibit the uptake of metal ions required for nitrogen fixation such as molybdenum, and that increasing sulfate concentrations negativelky affects algae such as cyanobacteria even at sulfate concentrations in estuarine (>8— 10 mM) and seawater (28mM) levels of sufate (see for example Marino et al, Hydrobiologia 500: pp. 277-293, 2004). Third, sulfate at high levels can often be taken up and reduced and sulfide, which is is toxic to photosynthesis because it attacks photosystem II (see for example Khanal et al., J. Envir. Engrg., 129(12); pp. 1104-1111, 2003). Fourth, high sulfate levels alter the osmotic pressure of the growth media, and many organisms cannot survive at such an elevated osmoticum. For example, it is well established that photosynthesis of algae is inhibited by hyperosmotic and salt stresses. See for example "Suppression of Quantum Yield of Photosystem II by Hyperosmotic Stress in Chlamydomonas reinhardtii" Plant Cell Physiol. 36: pp.1253-1258 (1995); and "The Effect of Osmotic and Ionic Stress on the Primary Processes of Photosynthesis in Dunaliella tertiolecta ", J. Exp. Bot. 1984 35(1): 18-27 (1984).
Microalgae can be grown in the presence of light. The number of photons striking a culture of microalgae cells can be manipulated, as well as other parameters such as the wavelength spectrum and ratio of darkdight hours per day. Microalgae can also be cultured in natural light, as well as simultaneous and/or alternating combinations of natural light and artificial light. For example, microalgae of the genus Chlorella be cultured under natural light during daylight hours and under artificial light during night hours.
The gas content of a photobioreactor can be manipulated. Part of the volume of a photobioreactor can contain gas rather than liquid. Gas inlets can be used to pump gases into the photobioreactor. Any gas can be pumped into a photobioreactor, including air, air/Cθ2 mixtures, noble gases such as argon and others. The rate of entry of gas into a photobioreactor can also be manipulated. Increasing gas flow into a photobioreactor increases the turbidity of a culture of microalgae. Placement of ports conveying gases into a photobioreactor can also affect the turbidity of a culture at a given gas flow rate. Air/CO2 mixtures can be modulated to generate different polysaccharide compositions by manipulating carbon flux. For example, air:CO2 mixtures of about 99.75% air:0.25% CO2, about 99.5% air:0.5% CO2, about 99.0% air: 1.00% CO2, about 98.0% air:2.0% CO2, about 97.0% air:3.0% CO2, about 96.0% air:4.0% CO2, and about 95.00% air:5.0% CO2 can be infused into a bioreactor or photobioreactor.
Microalgae cultures can also be subjected to mixing using devices such as spinning blades and propellers, rocking of a culture, stir bars, and other instruments.
B. Cell Culture Methods: Photobioreactors
Microalgae can be grown and maintained in closed photobioreactors made of different types of transparent or semitransparent material. Such material can include Plexiglas® enclosures, glass enclosures, bags bade from substances such as polyethylene, transparent or semitransparent pipes, and other materials. Microalgae can also be grown in open ponds.
Photobioreactors can have ports allowing entry of gases, solids, semisolids and liquids into the chamber containing the microalgae. Ports are usually attached to tubing or other means of conveying substances. Gas ports, for example, convey gases into the culture. Pumping gases into a photobioreactor can serve to both feed cells CO2 and other gases and to aerate the culture and therefore generate turbidity. The amount of turbidity of a culture varies as the number and position of gas ports is altered. For example, gas ports can be placed along the bottom of a cylindrical polyethylene bag. Microalgae grow faster when CO2 is added to air and bubbled into a photobioreactor. For example, a 5% CO2'.95% air mixture is infused into a photobioreactor containing cells of the genus Porphyridium (see for example Biotechnol Bioeng. 1998 Sep 20;59(6):705-13; textbook from office; US Patents 5,643,585 and 5,534,417; Lebeau, T., et. al. Appl. Microbiol Biotechnol (2003) 60:612-623; Muller-Fuega, A., Journal of Biotechnology 103 (2003 153-163; Muller-Fuega, A., Biotechnology and Bioengineering, Vol. 84, No. 5, December 5, 2003; Garcia- Sanchez, J.L., Biotechnology and Bioengineering, Vol. 84, No. 5, December 5, 2003; Garcia- Gonzales, M., Journal of Biotechnology, 115 (2005) 81-90. Molina Grima, E., Biotechnology Advances 20 (2003) 491-515).
Photobioreactors can be exposed to one or more light sources to provide microalgae with light as an energy source via light directed to a surface of the photobioreactor. Preferably the light source provides an intensity that is sufficient for the cells to grow, but not so intense as to cause oxidative damage or cause a photoinhibitive response. In some instances a light source has a wavelength range that mimics or approximately mimics the range of the sun. In other instances a different wavelength range is used. Photobioreactors can be placed outdoors or in a greenhouse or other facility that allows sunlight to strike the surface. Preferred photon intensities for species of the genus Porphyridium are between 50 and 300 uE m'2 s'1 (see for example Photosynth Res. 2005 Jun;84(l-3):21-7).
Photobioreactor preferably have one or more parts that allow media entry. It is not necessary that only one substance enter or leave a port. For example, a port can be used to flow culture media into the photobioreactor and then later can be used for sampling, gas entry, gas exit, or other purposes. In some instances a photobioreactor is filled with culture media at the beginning of a culture and no more growth media is infused after the culture is inoculated. In other words, the microalgal biomass is cultured in an aqueous medium for a period of time during which the microalgae reproduce and increase in number; however quantities of aqueous culture medium are not flowed through the photobioreactor throughout the time period. Thus in some embodiments, aqueous culture medium is not flowed through the photobioreactor after inoculation.
In other instances culture media can be flowed though the photobioreactor throughout the time period during which the microalgae reproduce and increase in number. In some instances media is infused into the photobioreactor after inoculation but before the cells reach a desired density. In other words, a turbulent flow regime of gas entry and media entry is not maintained for reproduction of microalgae until a desired increase in number of said microalgae has been achieved, but instead a parameter such as gas entry or media entry is altered before the cells reach a desired density.
Photobioreactors preferably have one or more ports that allow gas entry. Gas can serve to both provide nutrients such as CO2 as well as to provide turbulence in the culture media. Turbulence can be achieved by placing a gas entry port below the level of the aqueous culture media so that gas entering the photobioreactor bubbles to the surface of the culture. One or more gas exit ports allow gas to escape, thereby preventing pressure buildup in the photobioreactor. Preferably a gas exit port leads to a "one-way" valve that prevents contaminating microorganisms to enter the photobioreactor. In some instances cells are cultured in a photobioreactor for a period of time during which the microalgae reproduce and increase in number, however a turbulent flow regime with turbulent eddies predominantly throughout the culture media caused by gas entry is not maintained for all of the period of time. In other instances a turbulent flow regime with turbulent eddies predominantly throughout the culture media caused by gas entry can be maintained for all of the period of time during which the microalgae reproduce and increase in number. In some instances a predetermined range of ratios between the scale of the photobioreactor and the scale of eddies is not maintained for the period of time during which the microalgae reproduce and increase in number. In other instances such a range can be maintained.
Photobioreactors preferably have at least one port that can be used for sampling the culture. Preferably a sampling port can be used repeatedly without altering compromising the axenic nature of the culture. A sampling port can be configured with a valve or other device that allows the flow of sample to be stopped and started. Alternatively a sampling port can allow continuous sampling. Photobioreactors preferably have at least one port that allows inoculation of a culture. Such a port can also be used for other purposes such as media or gas entry.
Microalgae that produce polysaccharides can be cultured in photobioreactors. Microalgae that produce polysaccharide that is not attached to cells can be cultured for a period of time and then separated from the culture media and secreted polysaccharide by methods such as centrifugation and tangential flow filtration. Preferred organisms for culturing in photobioreactors to produce polysaccharides include Porphyridium sp., Porphyridium cruentum, Porphyridium purpureum, Porphyridium aerugineum, Rhodella maculata, Rhodella reticulata, Chlorella autotrophica, Chlorella stigmatophora, Chlorella capsulata, Achnanthes brevipes and Achnanthes longipes.
C. Non-Microalgal Polysaccharide Production
Organisms besides microalgae can be used to produce polysaccharides, such as lactic acid bacteria (see for example Stinglee, F., Molecular Microbiology (1999) 32(6), 1287-1295; Ruasjvladiedo, P., J. Dairy Sci. 88:843-856 (2005); Laws, A-, Biotechnology Advances 19 (2001) 597-625; Xanthan gum bacteria: Pollock, TJ., J. Ind. Microbiol Biotechnol., 1997 Aug; 19(2): 92-7.; Becker, A., Appl. Micrbiol. Bioltechnol. 1998 Aug;50(2):92-7; Garcia-Ochoa, F., Biotechnology Advances 18 (2000) 549-579., seaweed: Talarico, LB., et al., Antiviral Research 66 (2005) 103-110; Dussealt, J., et al., J Biomed Mater Res A., (2005) Novl; MeIo, F.R., J Biol Chem 279:20824-35 (2004)).
D. Ex Vivo Methods
Microalgae and other organisms can be manipulated to produce polysaccharide molecules that are not naturally produced by methods such as feeding cells with monosaccharides that are not produced by the cells (Nature. 2004 Aug 19;430(7002):873- 7). For example, species listed in Table 1 are grown according to the referenced growth protocols, with the additional step of adding to the culture media a fixed carbon source that is not in the culture media as published and referenced in Table 1 and is not produced by the cells in a detectable amount. In addition, such cells can first be transformed to contain a carbohydrate transporter, thus facilitating the entry of monosaccharides.
E. In vitro methods
Polysaccharides can be altered by enzymatic and chemical modification. For example, carbohydrate modifying enzymes can be added to a preparation of polysaccharide and allowed to catalyze reactions that alter the structure of the polysaccharide. Chemical methods can be used to, for example, modify the sulfation pattern of a polysaccharide (see for example Carbohydr. Polym. 63:75-80 (2000); Pomin VH., Glycobiology. 2005 Dec;15(12):1376-85; Naggi A., Semin Thromb Hemost. 2001 Oct;27(5):437-43 Review; Habuchi, O., Glycobiology. 1996 Jan;6(l);51-7; Chen, J., J. Biol. Chem. In press; Geresh., S et al., J. Biochem. Biophys. Methods 50 (2002) 179-187.).
F. Polysaccharide Purification Methods
Exopolysaccharides can be purified from microalgal cultures by various methods, including those disclosed herein.
Precipitation: For example, polysaccharides can be precipitated by adding compounds such as cetylpyridinium chloride, isopropanol, ethanol, or methanol to an aqueous solution containing a polysaccharide in solution. Pellets of precipitated polysaccharide can be washed and resuspended in water, buffers such as phosphate buffered saline or Tris, or other aqueous solutions (see for example Farias, W.R.L., et al., J. Biol. Chem. (2000) 275;(38)29299-29307; U.S. Patent No. 6,342,367; U.S. Patent No. 6,969,705). Dialysis: Polysaccharides can also be dialyzed to remove excess salt and other small molecules (see for example Gloaguen, V., et al., Carbohydr Res. 2004 Jan 2;339(l):97-103; Microbiol Immunol. 2000;44(5):395-400.).
Tangential Flow Filtration: Filtration can be used to concentrate polysaccharide and remove salts. For example, tangential flow filtration (TFF), also known as cross-flow filtration, can be used)). For a preferred filtration method see Geresh, Carb. Polym. 50; 183-189 (2002), which discusses use of a MaxCell A/G technologies 0.45uM hollow fiber filter. Also see for example Millipore Pellicon® devices, used with 10OkD, 30OkD, 1000 kD (catalog number P2C01MC01), 0.IuM (catalog number P2VVPPV01), 0.22uM (catalog number P2GVPPV01), and 0.45uM membranes (catalog number P2HVMPV01). It is preferred that the polysaccharides do not pass through the filter at a significant level. It is also preferred that polysaccharides do not adhere to the filter material. TFF can also be performed using hollow fiber filtration systems.
Non-limiting examples of tangential flow filtration include use of a filter with a pore size of at least about 0.1 micrometer, at least about 0.12 micrometer, at least about 0.14 micrometer, at least about 0.16 micrometer, at least about 0.18 micrometer, at least about 0.2 micrometer, at least about 0.22 micrometer, or at least about 0.45 micrometer. Preferred pore sizes of TFF allow contaminants to pass through but not polysaccharide molecules.
Tangential flow filtration may also be used to isolate and/or purify polysaccharides for further analysis or comparison. In some embodiments, the level of sulfation in polysaccharides from different cells or cell material may be analyzed or compared. In cases of intact cells or homogenized cell material, the cells or material may be resuspended in water or other suitable liquid to allow the release of polysaccharides into the liquid. The release may be augmented by agitation of the cells or material in the liquid, such as by vortexing, or by the use of an elevated temperature, such as a warm liquid. The released polysaccharides may be separated from the cells or insoluble cellular material by any convenient means, like centrifugation or filtration as non-limiting examples. The polysaccharide containing liquid may then be subjected to tangential flow filtration to isolate polysaccharides suitable for further analysis, such as combustion to determine the amount of sulfation in the polysaccharides. The amount of polysaccharide, or sulfation thereof, may be standardized against any known cellular metabolite or component, such as, but not limited to, the amount of DNA per gram of cells or cell material. In alternative embodiments, the polysaccharides from a cell extract may be analyzed as described above. Polysaccharides released into the extract, optionally with agitation and/or an elevated temperature as disclosed herein, may be separated from insoluble cellular material as described above. The soluble material may then be subjected to tangential flow filtration to isolate polysaccharides for further analysis or comparison.
In some embodiments, the amount or level of sulfation in the polysaccharides is analyzed and compared to the amount of sulfates used to culture the microalgae. Thus the amount or level of sulfation in the polysaccharides of cells grown at about 100 mM, about 200 mM, about 300 mM, about 400 mM, about 500 mM, about 600 mM, about 700 mM, or about 800 mM or higher, sulfate may be determined by routine and repetitive methods. Using sulfated exopolysaccharides as a non-limiting example, a disclosed polysaccharide may contain at least about 4.6% sulfur, at least about 4.75% sulfur, at least about 5.0% sulfur, at least about 5.25% sulfur, at least about 5.5% sulfur, at least about 5.75% sulfur, at least about 6.0% sulfur, at least about 6.25% sulfur, at least about 6.5% sulfur, at least about 6.75% sulfur, or at least about 7.0% sulfur by weight.
In further embodiments, the amount or level of sulfur by weight in the polysaccharides of a sample of cells or cell material may determined without knowledge of the amount of sulfate used to culture the cells. The amount or level of sulfur by weight may range from about 3.0 to about 7.0% as described above. In an alternative embodiment, the amount or level of sulfur may be used in a method to determine the amount of sulfates used to culture the cells and produce the cell material. The method may comprise comparison of the amount or level of sulfur, per unit of cellular metabolite or component, in a sample to that of cells grown in known amounts of sulfates. In further embodiments, the method may be used to identify cells, and so polysaccharides, prepared or obtained by methods disclosed herein. For example, homogenized Porphyrdium biomass may be incubated in distilled water, subjected to agitation to realease polysaccharide into solution, and cenytrifiged to remove solids. The resulting soluble polysaccharide can then be purified, concentrated, and diafiltered using methods disclosed in the Examples. The resulting ghigh purity polysaccharide may then be subjected to sulfur content analysis. The sulfur content alnalysis not only indicated the level of sulfation of the polysaccharide but also can indicate the level of sulfate under which the cells were cultured.
Ion Exchange Chromatography:_Anionic polysaccharides can be purified by anion exchange chromatography. (Jacobsson, I., Biochem J. 1979 Apr l;179(l):77-89; Karamanos, NK., Eur J Biochem. 1992 Mar. 1 ;204(2):553-60).
Protease Treatment: polysaccharides can be treated with proteases to degrade contaminating proteins. In some instances the contaminating proteins are attached, either covalently or noncovalently to polysaccharides. In other instances the polysaccharide molecules are in a preparation that also contains proteins. Proteases can be added to polysaccharide preparations containing proteins to degrade proteins (for example, the protease from Streptomyces griseus can be used (SigmaAldrich catalog number P5147). After digestion, the polysaccharide is preferably purified from residual proteins, peptide fragments, and amino acids. This purification can be accomplished, for example, by methods listed above such as dialysis, filtration, and precipitation.
Heat treatment can also be used to eliminate proteins in polysaccharide preparations (see for example Biotechnol Lett. 2005 Jan;27(l):13-8; FEMS Immunol Med Microbiol. 2004 Oct 1;42(2): 155-66; Carbohydr Res. 2000 Sep 8;328(2):199-207; J Biomed Mater Res. 1999;48(2):l l l-6.; Carbohydr Res. 1990 Oct 15;207(l):101-20; ).
The invention thus includes production of an exopolysaccharide comprising separating the exopolysaccharide from contaminants after proteins attached to the exopolysaccharide have been degraded or destroyed. The proteins may be those attached to the exopolysaccharide during culture of a microalgal cell in media, which is first separated from the cells prior to proteolysis or protease treatment. The cells may be those of the genus Porphyridium as a non-limiting example.
In one non-limiting example, a method of producing an exopolysaccharide is provided wherein the method comprises culturing cells of the genus Porphyridium; separating cells from culture media; destroying protein attached to the exopolysaccharide present in the culture media; and separating the exopolysaccharide from contaminants. In some methods, the contaminant(s) are selected from amino acids, peptides, proteases, protein fragments, and salts. In other methods, the contaminant is selected from NaCl, MgSO4, MgCl2, CaCl2, KNO3, KH2PO4, NaHCO3, Tris, ZnCl2, H3BO3, CoCl2, CuCl2, MnCl2, (NH4) 6Mo7O24, FeC13 and EDTA.
Drying Methods: After purification of methods such as those above, polysaccharides can be dried using methods such as lyophilization and heat drying (see for example Shastry, S., Brazilian Journal of Microbiology (2005) 36:57-62; Matthews KH.Jnt J Pharm. 2005 Jan 31;289(l-2):51-62. Epub 2004 Dec 30; Gloaguen, V., et al., Carbohydr Res. 2004 Jan 2;339(l):97-103).
Tray dryers accept moist solid on trays. Hot air (or nitrogen) is circulated to dry. Shelf dryers can also employ reduced pressure or vacuum to dry at room temperature when products are temperature sensitive and are similar to a freeze-drier but less costly to use and can be easily scaled-up. Spray dryers are relatively simple in operation, which accept feed in fluid state and convert it into a dried particulate form by spraying the fluid into a hot drying medium.
Rotary dryers operate by continuously feeding wet material, which is dried by contact with heated air, while being transported along the interior of a rotating cylinder, with the rotating shell acting as the conveying device and stirrer.
Spin flash dryers are used for drying of wet cake, slurry, or paste which is normally difficult to dry in other dryers. The material is fed by a screw feeder through a variable speed drive into the vertical drying chamber where it is heated by air and at the same time disintegrated by a specially designed disintegrator. The heating of air may be direct or indirect depending upon the application. The dry powder is collected through a cyclone separator/bag filter or with a combination of both.
Whole Cell Extraction: Intracellular polysaccharides and cell wall polysaccharides can be purified from whole cell mass (see form example U.S. Patent 4,992,540; U.S. Patent 4,810,646; J Sietsma JH., et al., Gen Microbiol. 1981 Jul;125(l):209-12; Fleet GH, Manners DJ., J Gen Microbiol. 1976 May;94(l): 180-92).
G. Microalgae Homogenization Methods
A pressure disrupter pumps of a slurry through a restricted orifice valve. High pressure (up to 1500 bar) is applied, followed by an instant expansion through an exiting nozzle. Cell disruption is accomplished by three different mechanisms: impingement on the valve, high liquid shear in the orifice, and sudden pressure drop upon discharge, causing an explosion of the cell. The method is applied mainly for the release of intracellular molecules. According to Hetherington et al., cell disruption (and consequently the rate of protein release) is a first-order process, described by the relation: log(Rm/(Rm-R)) = K N P 72.9. R is the amount of soluble protein; Rm is the maximum amount of soluble protein K is the temperature dependent rate constant; N is the number of passes through the homogenizer (which represents the residence time). P is the operating pressure.
In a ball mill, cells are agitated in suspension with small abrasive particles. Cells break because of shear forces, grinding between beads, and collisions with beads. The beads disrupt the cells to release biomolecules. The kinetics of biomolecule release by this method is also a first-order process. In some embodiments, cell containing samples are cryogenically ball milled in a planetary ball mill (Retsch, PMlOO) at 10-80 g batch size. The powder is placed in a grinding bowl with eight to ten %-inch-diameter stainless steel balls. The sample is cooled repeatedly with liquid nitrogen. The material is milled at 400- 550 rpm for 30 to 60 min. The final product is dried in a desiccator overnight.
Another widely applied method is the cell lysis with high frequency sound that is produced electronically and transported through a metallic tip to an appropriately concentrated cellular suspension, ie: sonication. The concept of ultrasonic disruption is based on the creation of cavities in cell suspension.
In some embodiments the invention comprises polysaccharides from a species of the genus Porphyridium, optionally sulfated at a level of 4.7 or higher, that have been reduced in molecular weight through milling or shearing such as, for example, sonication or ball milling.
Blending (high speed or Waring), the french press, or even centrifugation in case of weak cell walls, also disrupt the cells by using the same concepts.
Cells can also be ground after drying in devices such as a colloid mill.
In optional embodiments, the invention includes a method of preparing a dry homogenate by use of lyophilization. As a non-limiting example, a cell containing material is lyophilized prior to homogenization, such as by grinding or milling. In some cases, the cell containing material comprises both cells (biomass) and soluble polysaccharides in the culture medium. In other embodiments, only biomass is used. To prepare a dry material, the cell-containing material is optionally dried (at least partially) or otherwise concentrated before subsequent lyophilization. The lyophilized material may be homogenized to produce a homogenate comprising a powder or particles of a subcellular size range. Non-limiting examples include particles that are less than 800 microns in diameter, less than 200 microns in diameter, less than 100 microns in diameter, less than 50 microns in diameter, less than 10 microns in diameter, less than 5 microns in diameter, and less than 1 micron in diameter. In some embodiments the average particle size of a composition disclosed herein is less than 800 microns in diameter, less than 200 microns in diameter, less than 100 microns in diameter, less than 50 microns in diameter, less than 10 microns in diameter, less than 5 microns in diameter, and less than 1 micron in diameter.
Without being bound by theory, and offered to improve the understanding of the invention, the use of homogenized cells (or biomass) is believed to improve the bioavailability of polysaccharides after administration to a mammal as described herein. As a non-limiting example, the homogenized material may improve exposure of the polysaccharides to solvent and subsequent uptake in the digestive tract of a mammal. Alternatively, the homogenization process may fragment the polysaccharides under certain conditions, such as to produce fragments of sulfated polysaccharides, which increase the total activity available in the homogentate. A further alternative is that the homogenization process releases intracellular polysaccharides which add to the total activity in the homogenate. While none of these proposed mechanisms may be responsible for the improved activity seen with homogenates of cells, a combination of two or more of the above mechanisms also may be responsible for the improved activity.
The use of cell material, such as a homogenate, appears to be contrary to earlier reports of improved effects seen with the use of purified polysaccharides compared to unpurified biomass (see Dvir et al. Br. J. Nutri. (2000) 84:469-476; and Ginzberg et al. J. Appl. Phycology (2000) 12:325-330). These earlier reports appear to suggest that biomass is not as effective as purified polysaccharide. But as evident by data disclosed herein, homogenized biomass is effective at reducing total serum cholesterol (TSC), and more so than intact biomass. For example, there is an approximately 8% decrease in TSC between homogenate and intact biomass administered at the same dosage (see Figure 10 herein, groups 2 vs. 6). In addition, a biomass homogenate at a 5% or 10% diet reduced TSC to a much greater extent (approximately 39% and 54% respectively) than non- homogenized biomass administered at much higher dosages as discussed in the literature (22% reduction in TSC observed for a 19% diet of unpurified biomass- see Dvir et al. Br. J. Nutri. (2000) 84:469-476). As such it was a surprising result to see that lower dosages of a biomass homogenate generate such a significant reduction in TSC.
There is an approximately 8% decrease in total serum cholesterol between homogenized and intact biomass (see figure 10, groups 2 vs. 6). In addition, homogenized biomass at a 5% and 10% diet reduced total serum cholesterol to a much greater extent (approximately 39% and 54% respectively) than non-homogenized biomass administered at much higher dosages discussed in the literature (22% reduction in total serum cholesterol for a 19% diet of intact biomass- see Dvir et al. Br. J. Nutri. (2000) 84:469-476). As such it was a surprising result to see that lower dosages of biomass, when processed according to the methods disclosed herein, provide far greater reduction in total serum cholesterol.
Because the percentage of polysaccharide as a function of the dry weight of a microalgae cell can frequently be in excess of 50%, microalgae cell homogenates can be considered partially pure polysaccharide compositions. Cell disruption aids in increasing the amount of solvent-accessible polysaccharide by breaking apart cell walls that are largely composed of polysaccharide. Homogenizatioπ as described herein can increase the amount of solvent-available polysaccharide significantly. For example, homogenization can increase the amount of solvent-available polysaccharide by at least a factor of 0.25, at least a factor, of 0.5, at least a factor of 1, at least a factor of 2, at least a factor of 3, at least a factor of 4, at least a factor of 5, at least a factor of 8, at least a factor of 10, at least a factor of 15, at least a factor of 20, at least a factor of 25, and at least a factor of 30 or more compared to the amount of solvent- available polysaccharide in an identical or similar quantity of non-homogenized cells of the same type. One way of determining a quantity of cells sufficient to generate a given quantity of homogenate is to measure the amount of a compound in the homogenate and calculate the gram quantity of cells required to generate this amount of the compound using known data for the amount of the compound per gram mass of cells. The quantity of many such compounds per gram of particular microalgae cells are known. For examples, see Figure 9. Given a certain quantity of a compound in a composition, the skilled artisan can determine the number of grams of intact cells necessary to generate the observed amount of the compound. The number of grams of microalgae cells present in the composition can then be used to determine if the composition contains at least a certain amount of solvent- available polysaccharide sufficient to indicate whether or not the composition contains homogenized cells, such as for example five times the amount of solvent-available polysaccharide present in a similar or identical quantity of unhomogenized cells.
H. Analysis Methods
Assays for detecting polysaccharides can be used to quantitate starting polysaccharide concentration, measure yield during purification, calculate density of secreted polysaccharide in a photobioreactor, measure polysaccharide concentration in a finished product, and other purposes..
The phenol: sulfuric acid assay detects carbohydrates (see Hellebust, Handbook of Phycological Methods, Cambridge University Press, 1978; and Cuesta G., et al., J Microbiol Methods. 2003 Jan;52(l):69-73). The 1,6 dimethylmethylene blue assay detects anionic polysaccharides, (see for example Braz J Med Biol Res. 1999 May;32(5):545-50; Clin Chem. 1986 Nov;32(ll):2073-6).
Polysaccharides can also be analyzed through methods such as HPLC, size exclusion chromatography, and anion exchange chromatography (see for example Prosky L, Asp N, Schweizer TF, DeVries JW & Furda I (1988) Determination of insoluble, soluble and total dietary fiber in food and food products: Interlaboratory study. Journal of the Association of Official Analytical Chemists 71, 1017±1023; Int J Biol Macromol. 2003 Nov;33(l-3):9-18)
Polysaccharides can also be detected using gel electrophoresis (see for example Anal Biochem. 2003 Oct 15;321(2):174-82; Anal Biochem. 2002 Jan l;300(l):53-68).
Monosaccharide analysis of polysaccharides can be performed by combined gas chromatography/mass spectrometry (GC/MS) of the per-O-trimethylsilyl (TMS) derivatives of the monosaccharide methyl glycosides produced from the sample by acidic methanolysis (see Merkle and Poppe (1994) Methods Enzymol. 230: 1-15; York, et al. (1985) Methods Enzymol. 118:3-40).
Ill Compositions
A. General
Compositions of the invention include a microalgal polysaccharide or homogenate as described herein. In embodiments relating to polysaccharides, including exopolysaccharides, the composition may comprise a homogenous or a heterogeneous population of polysaccharide molecules, including sulfated polysaccharides as non-limiting embodiments. Non-limiting examples of homogenous populations include those containing a single type of polysaccharide molecule, such as that with the same structure and molecular weight. Non- limiting examples of heterogeneous populations include those containing more than one type of polysaccharide molecule, such as a mixture of polysaccharides having a molecular weight (MW) within a range or a MW above or below a MW value. For example, the Porphyridium sp. exopolysaccharide is typically produced in a range of sizes from 3-5 million Daltons. Of course a polysaccharide containing composition of the invention may be optionally protease treated, or reduced in the amount of protein, as described above.
In some embodiments, a composition of the invention may comprise one or more polysaccharides produced by microalgae that have not been recombinantly modified. The microalgae may be those which are naturally occurring or those which have been maintained in culture in the absence of alteration by recombinant DNA techniques or genetic engineering.
In other embodiments, the polysaccharides are those from modified microalgae, such as, but not limited to, microalgae modified by recombinant techniques. Non-limiting examples of such techniques include introduction and/or expression of an exogenous nucleic acid sequence encoding a gene product; genetic manipulation to decrease or inhibit expression of an endogenous microalgal gene product; and/or genetic manipulation to increase expression of an endogenous microalgal gene product. The invention contemplates recombinant modification of the various microalgae species described herein. In some embodiments, the microalgae is from the genus Porphyridium.
Polysaccharides provided by the invention that are produced by genetically modified microalgae or microalgae that are provided with an exogenous carbon source can be distinct from those produced by microalgae cultured in minimal growth media under photoautotrophic conditions (ie: in the absence of a fixed carbon source) at least in that they contain a different monosaccharide content relative to polysaccharides from unmodified microalgae or microalgae cultured in minimal growth media under photoautotrophic conditions. Non-limiting examples include polysaccharides having an increased amount of arabinose (Ara), rhamnose (Rha), fucose (Fuc), xylose (XyI), glucuronic acid (GIcA), galacturonic acid (GaIA), mannose (Man), galactose (Gal), glucose (GIc), N-acetyl galactosamine (GaINAc), N-acetyl glucosamine (GIcNAc), and/or N-acety] neuraminic acid (NANA)5 per unit mass (or per mole) of polysaccharide, relative to polysaccharides from either non-genetically modified microalgae or microalgae cultured photoautotrophically. An increased amount of a monosaccharide may also be expressed in terms of an increase relative to other monosaccharides rather than relative to the unit mass, or mole, of polysaccharide. An example of genetic modification leading to production of modified polysaccharides is transforming a microalgae with a carbohydrate transporter gene, and culruring a transformant in the presence of a monosaccharide which is transported into the cell from the culture media by the carbohydrate transporter protein encoded by the carbohydrate transporter gene. In some instances the culture can be in the dark, where the monosaccharide, such as glucose, is used as the sole energy source for the cell. In other instances the culture is in the light, where the cells undergo photosynthesis and therefore produce monosaccharides such as glucose in the chloroplast and transport the monosaccharides into the cytoplasm, while additional exogenously provided monosaccharides are transported into the cell by the carbohydrate transporter protein. In both instances monosaccharides from the cytoplasm are transported into the endoplasmic reticulum, where polysaccharide synthesis occurs. Novel polysaccharides produced by non- genetically engineered microalgae can also be generated by nutritional manipulation, ie: exogenously providing carbohydrates in the culture media that are taken up through endogenous transport mechanisms. Uptake of the exogenously provided carbohydrates can be induced, for example, by culruring the cells in the dark, thereby forcing the cells to utilize the exogenously provided carbon source. For example, Porphyridium cells cultured in the presence of 7% glycerol in the dark produce a novel polysaccharide because the intracellular carbon flux under these nutritionally manipulated conditions is different from that under photosynthetic conditions. Insertion of carbohydrate transporter genes into microalgae facilitates, but is not strictly necessary for, polysaccharide structure manipulation because expression of such genes can significantly increase the concentration of a particular monosaccharide in the cytoplasm of the cell. Many carbohydrate transporter genes encode proteins that transport more than one monosaccharide, albeit with different affinities for different monosaccharides (see for example Biochimica et Biophysica Acta 1465 (2000) 263-274). In some instances a microalgae species can be transformed with a carbohydrate transporter gene and placed under different nutritional conditions, wherein one set of conditions includes the presence of exogenously provided galactose, and the other set of conditions includes the presence of exogenously provided xylose, and the transgenic species produces structurally distinct polysaccharides under the two conditions. By altering the identity and concentration of monosaccharides in the cytoplasm of the microalgae, through genetic and/or nutritional manipulation, the invention provides novel polysaccharides. Nutritional manipulation can also be performed, for example, by culturing the microalgae in the presence of high amounts of sulfate, as described herein. In some instances nutritional manipulation includes addition of one or more exogenously provided carbon sources as well as one or more other non-carbohydrate culture component, such as 5OmM MgSO4.
In some embodiments, the increase in one or more of the above listed monosaccharides in a polysaccharide may be from below to above detectable levels and/or by at least about 5%, to at least about 2000%, relative to a polysaccharide produced from the same microalgae in the absence of genetic or nutritional manipulation. Therefore an increase in one or more of the above monosaccharides, or other carbohydrates listed in Tables 2 or 3S by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 100%, at least about 105%, at least about 110%, at least about 150%, at least about 200%, at least about 250%, at least about 300%, at least about 350%, at least about 400%, at least about 450%, at least about 500%, at least about 550%, at least about 600%, at least about 650%, at least about 700%, at least about 750%, at least about 800%, at least about 850%, at least about 900%, at least about 1000%, at least about 1100%, at least about 1200%, at least about 1300%, at least about 1400%, at least about 1500%, at least about 1600%, at least about 1700%, at least about 1800%, or at least about 1900%, or more, may be used in the practice of the invention.
In cases wherein the polysaccharides from unmodified microalgae do not contain one or more of the above monosaccharides, the presence of the monosaccharide in a microalgal polysaccharide indicates the presence of a polysaccharide distinct from that in unmodified microalgae. Thus using particular strains of Po rphyridium sp. and Porphyridium cruentum as non-limiting examples, the invention includes modification of these microalgae to incorporate arabinose and/or fucose, because polysaccharides from two strains of these species do not contain detectable amounts of these monosaccharides (see Example 5 herein), hi another non-limiting example, the modification of Porphyridium sp. to produce polysaccharides containing a detectable amount of glucuronic acid, galacturonic acid, or N-acetyl galactosamine, or more than a trace amount of N-acetyl glucosamine, is specifically included in the instant disclosure. In a further non-limiting example, the modification of Porphyridium cruentum to produce polysaccharides containing a detectable amount of rhamnose, mannose, or N-acetyl neuraminic acid, or more than a trace amount of N-acetyl-glucosamine, is also specifically included in the instant disclosure.
Put more generally, the invention includes a method of producing a polysaccharide comprising culturing a microalgae cell in the presence of at least about 0.01 micromolar of an exogenously provided fixed carbon compound, wherein the compound is incorporated into the polysaccharide produced by the cell. In some embodiments, the compound is selected from Table 2 or 3. The cells may optionally be selected from the species listed in Table 1, and cultured by modification, using the methods disclosed herein, or the culture conditions also lusted in Table 1.
The methods may also be considered a method of producing a glycopolymer by culturing a transgenic microalgal cell in the presence of at least one monosaccharide, wherein the monosaccharide is transported by the transporter into the cell and is incorporated into a microalgal polysaccharide.
In some embodiments, the cell is selected from Table 1, such as where the cell is of the genus Porphyridium, as a non-limiting example. In some cases, the cell is selected from Porphyridium sp. and Porphyridium cruentum. Embodiments include those wherein the polysaccharide is enriched for the at least one monosaccharide compared to an endogenous polysaccharide produced by a non-transgenic cell of the same species. The monosaccharide may be selected from Arabinose, Fructose, Galactose, Glucose, Mannose, Xylose, Glucuronic acid, Glucosamine, Galactosamine, Rhamnose and N-acetyl glucosamine.
These methods of the invention are facilitated by use of a transgenic cell expressing a sugar transporter, optionally wherein the transporter has a lower Kn, for glucose than at least one monosaccharide selected from the group consisting of galactose, xylose, glucuronic acid, mannose, and rhamnose. In other embodiments, the transporter has a lower Km for galactose than at least one monosaccharide selected from the group consisting of glucose, xylose, glucuronic acid, mannose, and rhamnose. In additional embodiments, the transporter has a lower Km for xylose than at least one monosaccharide selected from the group consisting of glucose, galactose, glucuronic acid, mannose, and rhamnose. In further embodiments, the transporter has a lower Km for glucuronic acid than at least one monosaccharide selected from the group consisting of glucose, galactose, xylose, mannose, and rhamnose. In yet additional embodiments, the transporter has a lower Km for mannose than at least one monosaccharide selected from the group consisting of glucose, galactose, xylose, glucuronic acid, and rhamnose. In yet further embodiments, the transporter has a lower Km for rhamnose than at least one monosaccharide selected from the group consisting of glucose, galactose, xylose, glucuronic acid, and mannose. Manipulation of the concentration and identity of monosaccharides provided in the culture media, combined with use of transporters that have a different Km for different monosaccharides, provides novel polysaccharides. These general methods can also be used in cells other than microalgae, for example, bacteria that produce polysaccharides.
In alternative embodiments, the cell is cultured in the presence of at least two monosaccharides, both of which are transporter by the transporter. In some cases, the two monosaccharides are any two selected from glucose, galactose, xylose, glucuronic acid, rhamnose and mannose.
In one non-limiting example, the method comprises providing a transgenic cell containing a recombinant gene encoding a monosaccharide transporter; and culturing the cell in the presence of at least one monosaccharide, wherein the monosaccharide is transported by the transporter into the cell and is incorporated into a polysaccharide of the cell. It is pointed out that transportation of a monosaccharide from the media into a microalgal cell allows for the monosaccharide to be used as an energy source, as disclosed below, and for the monosaccharide to be transported into the endoplasmic reticulum (ER) by cellular transporters. In the ER, polysaccharide production and glycosylation, occurs such that in the presence of exogenously provided monosaccharides, the sugar content of the microalgal polysaccharides change.
In some aspects, the invention includes a novel microalgal polysaccharide, such as from microalgae of the genus Porphyridium, comprising detectable amounts of xylose, glucose, and galactose wherein the molar amount of one or more of these three monosaccharides in the polysaccharide is not present in a polysaccharide of Porphyridium that is not genetically or nutritionally modified. An example of a non-nutritionally and non- genetically modified Porphyridium polysaccharide can be found, for example, in Jones R., Journal of Cellular Comparative Physiology 60; 61-64 (1962). In some embodiments, the amount of glucose, in the polysaccharide, is at least about 65% of the molar amount of galactose in the same polysaccharide. In other embodiments, glucose is at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 100%, at least about 105%, at least about 110%, at least about 120%, at least about 130%, at least about 140%, at least about 150%, at least about 200%, at least about 250%, at least about 300%, at least about 350%, at least about 400%, at least about 450%, at least about 500%, or more, of the molar amount of galactose in the polysaccharide. Further embodiments of the invention include those wherein the amount of glucose in a microalgal polysaccharide is equal to, or approximately equal to, the amount of galactose (such that the amount of glucose is about 100% of the amount of galactose). Moreover, the invention includes microalgal polysaccharides wherein the amount of glucose is more than the amount of galactose.
Alternatively, the amount of glucose, in the polysaccharide, is less than about 65% of the molar amount of galactose in the same polysaccharide. The invention thus provides for polysaccharides wherein the amount of glucose is less than about 60%, less than about 55%, less than about 50%, less than about 45%, less than about 40%, less than about 35%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, or less than about 5% of the molar amount of galactose in the polysaccharide.
In other aspects, the invention includes a microalgal polysaccharide, such as from microalgae of the genus Porphyridium, comprising detectable amounts of xylose, glucose, galactose, mannose, and rhamnose, wherein the molar amount of one or more of these five monosaccharides in the polysaccharide is not present in a polysaccharide of non-genetically modified and/or non-nutritionally modified microalgae. In some embodiments, the amount of rhamnose in the polysaccharide is at least about 100% of the molar amount of mannose in the same polysaccharide. In other embodiments, rhamnose is at least about 110%, at least about 120%, at least about 130%, at least about 140%, at least about 150%, at least about 200%, at least about 250%, at least about 300%, at least about 350%, at least about 400%, at least about 450%, or at least about 500%, or more, of the molar amount of mannose in the polysaccharide. Further embodiments of the invention include those wherein the amount of rhamnose in a microalgal polysaccharide is more than the amount of mannose on a molar basis.
Alternatively, the amount of rhamnose, in the polysaccharide, is less than about 75% of the molar amount of mannose in the same polysaccharide. The invention thus provides for polysaccharides wherein the amount of rhamnose is less than about 70%, less than about 65%, less than about 60%, less than about 55%, less than about 50%, less than about 45%, less than about 40%, less than about 35%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, or less than about 5% of the molar amount of mannose in the polysaccharide.
In additional aspects, the invention includes a microalgal polysaccharide, such as from microalgae of the genus Porphyridium, comprising detectable amounts of xylose, glucose, galactose, mannose, and rhamnose, wherein the amount of mannose, in the polysaccharide, is at least about 130% of the molar amount of rhamnose in the same polysaccharide. In other embodiments, mannose is at least about 140%, at least about 150%, at least about 200%, at least about 250%, at least about 300%, at least about 350%, at least about 400%, at least about 450%, or at least about 500%, or more, of the molar amount of rhamnose in the polysaccharide.
Alternatively, the amount of mannose, in the polysaccharide, is equal to or less than the molar amount of rhamnose in the same polysaccharide. The invention thus provides for polysaccharides wherein the amount of mannose is less than about 95%, less than about 90%, less than about 85%, less than about 80%, less than about 75%, less than about 70%, less than about 65%, less than about 60%, less than about 60%, less than about 55%, less than about 50%, less than about 45%, less than about 40%, less than about 35%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, or less than about 5% of the molar amount of rhamnose in the polysaccharide.
In further aspects, the invention includes a microalgal polysaccharide, such as from microalgae of the genus Porphyridium, comprising detectable amounts of xylose, glucose, and galactose, wherein the amount of galactose in the polysaccharide, is at least about 100% of the molar amount of xylose in the same polysaccharide. In other embodiments, rhamnose is at least about 105%, at least about 110%, at least about 120%, at least about 130%, at least about 140%, at least about 150%, at least about 200%, at least about 250%, at least about 300%, at least about 350%, at least about 400%, at least about 450%, or at least about 500%, or more, of the molar amount of mannose in the polysaccharide. Further embodiments of the invention include those wherein the amount of galactose in a microalgal polysaccharide is more than the amount of xylose on a molar basis.
Alternatively, the amount of galactose, in the polysaccharide, is less than about 55% of the molar amount of xylose in the same polysaccharide. The invention thus provides for polysaccharides wherein the amount of galactose is less than about 50%, less than about 45%, less than about 40%, less than about 35%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, or less than about 5% of the molar amount of xylose in the polysaccharide.
In yet additional aspects, the invention includes a microalgal polysaccharide, such as from microalgae of the genus Porphyridium, comprising detectable amounts of xylose, glucose, glucuronic acid and galactose, wherein the molar amount of one or more of these five monosaccharides in the polysaccharide is not present in a polysaccharide of unmodified microalgae. In some embodiments, the amount of glucuronic acid, in the polysaccharide, is at least about 50% of the molar amount of glucose in the same polysaccharide. In other embodiments, glucuronic acid is at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 100%, at least about 105%, at least about 110%, at least about 120%, at least about 130%, at least about 140%, at least about 150%, at least about 200%, at least about 250%, at least about 300%, at least about 350%, at least about 400%, at least about 450%, or at least about 500%, or more, of the molar amount of glucose in the polysaccharide. Further embodiments of the invention include those wherein the amount of glucuronic acid in a microalgal polysaccharide is more than the amount of glucose on a molar basis.
In other embodiments, the exopolysaccharide, or cell homogenate polysaccharide, comprises glucose and galactose wherein the molar amount of glucose in the exopolysaccharide, or cell homogenate polysaccharide, is at least about 55% of the molar amount of galactose in the exopolysaccharide or polysaccharide. Alternatively, the molar amount of glucose in the exopolysaccharide, or cell homogenate polysaccharide, is at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, or at least about 100% of the molar amount of galactose in the exopolysaccharide or polysaccharide.
In yet further aspects, the invention includes a microalgal polysaccharide, such as from microalgae of the genus Porphyridium, comprising detectable amounts of xylose, glucose, glucuronic acid, galactose, at least one monosaccharide selected from arabinose, fucose, N-acetyl galactosamine, and N-acetyl neuraminic acid, or any combination of two or more of these four monosaccharides.
Embodiments of the invention include a composition comprising a intact red microalgae cells or a homogenate of red microalgae cells. Non-limiting examples include a powder or particles as disclosed herein. In some embodiments, the composition further includes a carrier suitable for consumption by a mammal, such as a human. Therefore, embodiments of the invention include a food product, such as a human food product, comprising a homogenate of red microalgae cells. In alternative embodiments, the composition may be a capsule or tablet comprising the homogenate.
Additional embodiments of the invention include compositions wherein the red microalgae cells are of the genus Porphyridium. Non-limiting examples include red microalgae cells that are Porphyridium sp. UTEX 637, or a strain derived from Porphyridium sp. UTEX 637; Porphyridium cruentum UTEX 161, or a strain derived from Porphyridium cruentum UTEX 161; Porphyridium aerugineum, or a strain derived from Porphyridium aerugineum; Porphyridium sordidum, or a strain derived from Porphyridium sordidum; or Porphyridium purpureum, or a strain derived from Porphyridium purpureum.
Further embodiments include compositions wherein the homogenate comprises a sulfated exopolysaccharide, secreted by the red microalgae cells, wherein the polysaccharide in purified form contains at least about 4.6% sulfur, at least about 4.75% sulfur, at least about 5.0% sulfur, at least about 5.25% sulfur, at least about 5.5% sulfur, at least about 5.75% sulfur, at least about 6.0% sulfur, at least about 6.25% sulfur, at least about 6.5% sulfur, at least about 6.75% sulfur, or at least about 7.0% sulfur by weight.
Additional embodiments include compositions wherein the homogenate comprises at least about two, at least about three, at least about four, at least about five, at least about six, at least about seven, at least about eight, at least about nine, at least about 10, at least about 12, at least about 14, at least about 16, at least about 18, or at least about 20 times the amount of solvent-available polysaccharide compared to the quantity of unhomogenized cells (biomass) used to form the homogenate. Stated differently, the homogenate contains from about two, to about 20 or more, times the amount or level of solvent-available polysaccharide compared to the unhomogenized cells needed to generate the microalgal cell homogenate.
Embodiments of the invention also include compositions comprising combinations of the above. In some cases, the composition comprises a homogenate with a sulfated exopolysaccharide secreted by red microalgae cells that contains at least 3.0%, or at least 4.75%, sulfur by weight, and the homogenate contains at least two times the amount of solvent-available polysaccharide as present in a quantity of unhomogenized cells needed to generate the microalgal cell homogenate.
In some embodiments the invention includes mixtures of polysaccharides from Porphyridium as described herein and polysaccharides from a species of Chlorella, such as Chlorella marina, Chlorella salina, Chlorella pyrenoidosa, Chlorella vulgaris, Chlorella anitrata, Chlorella antarctica, Chlorella autotrophica, Chlorella regularis, among others (see World Catalog of Algae, 2.sup.nd Edition, pages 58-74; Miyachi et al. (Eds); 1989; Japan Scientific Societies Press Chlorella minutissima and Chlorella pyrenoidosa, as described in US Patents 6,974,576, 6,551,596, 4,831,020, 4,533,548 and 6,977,076.
B. Cholesterol Lowering Compositions
Polysaccharides from microalgae can be formulated for ingestion to achieve a hypocholesterolemic effect. For example, the secreted polysaccharide from Porphyridium sp. can be formulated for administration as a cholesterol lowering agent. Secreted polysaccharides from Porphyridium cruentum, Porphyridium purpureum, Porphyridium aerugineum, Rhodella maculata, Rhodella reticulata, Chlorella autotrophica, Chlorella stigmatophora, Chlorella capsulata, Achnanthes brevipes and Achnanthes longipes can also be formulated for administration as a cholesterol lowering agent. These microalgae are cultured, for example, in photobioreactors in the presence of light, more preferably in the presence of strong light such as 175 μmol photons per square meter per second, for a period of time sufficient for the cells to secrete polysaccharide molecules. Some species, such as those of Chlorella and Porphyridium, can also be cultured in the absence of light and in the presence of a fixed carbon source. In some embodiments, the polysaccharides or polysaccharide material will be from a Porphyridium species, such as one that has been subject to genetic and/or nutritional manipulation to produce polysaccharides with altered monosaccharide content and/or altered sulfation.
Patients in need of cholesterol lowering polysaccharide agents such as polysaccharides are preferably those with total cholesterol above 200 mg/dL, those with LDL Cholesterol above 130 mg/dL, those with HDL Cholesterol less than 40 mg/dL, and those with triglycerides above 150 mg/dL.
The invention also comprises administering to a patient described herein a combination of an algal polysaccharide such as that from a cell of the genus Porphyridium and another compound such as a plant phytosterol or a statin such as Pravachol®, Mevacor®, Zocor®, Lescol®, Lipitor®, Baycol®, Crestor®, and Advicor®. The invention also comprises a method of reducing the side effects of a statin drug comprising lowering the dosage of a statin and administering a polysaccharide produced from microalgae, such as for example the polysaccharide from a cell of the genus Porphyridium. Side effects from statins include nausea, irritability and short temper, hostility, homicidal impulses, loss of mental clarity, amnesia, kidney failure, diarrhea, muscle aching and weakness, tingling or cramping in the legs, inability to walk, sleeping problems, constipation, impaired muscle formation, erectile dysfunction, temperature regulation problems, nerve damage, mental confusion, liver damage and abnormalities, neuropathy, and destruction of CoQlO. The invention also includes administering a polysaccharide produced from microalgae, such as for example the polysaccharide from a cell of the genus Porphyridium, to a patient with total cholesterol of 240 mg/dL or more; to a patient with LDL Cholesterol of 130 to 159 mg/dL, 160 to 189 mg/dL, and 190 mg/dL or higher; and to a patient with triglycerides of 150 to 199 mg/dL, or 200 mg/dL or higher.
In one embodiment, cells of the genus Porphyridium are harvested from culture and homogenized to form a composition for administration to lower cholesterol. Homogenization of the cells provides an increased level of bioavailability of the cell wall polysaccharide compared to intact cells. Homogenization can be performed by methods such as sonication, jet milling, colloid milling, wet grinding, dry grinding, and other methods. A preferred composition for cholesterol reduction is homogenized Porphyridium, wherein the average particle size is less then 300 microns, more preferably less than 200 microns, more preferably less than 100 microns, more preferably less than 50 microns, more preferably less than 25 microns, and more preferably less than 10 microns. In some embodiments the cells are dried before grinding, while in other embodiments homogenization is performed on wet cells, such as sonication. Homogenization of microalgae to increase bioavailability of cell wall polysaccharides can be performed to produce homogenates, also referred to herein as polysaccharide material, of any microalgae, including species from Table 1. Polysaccharides of the invention may be formulated as a composition for oral consumption, as in a dietary supplement as a non- limiting example. The formulation may be in solid or liquid form. For example, purified lyophilized polysaccharide can be formulated in capsules or tablets. Conventional methods for the preparation of capsules or tablets are known to the skilled person. The methods may include use of pharmaceutically . acceptable excipients such as binding agents, fillers, disintegrants, or wetting agents, sweeteners, including, pregelatinised maize starch, polyvinylpyrrolidone, hydroxypropyl methylcellulose, fillers, lactose, microcrystalline cellulose, calcium hydrogen phosphate, lubricants, magnesium stearate, talc, silica, potato starch or sodium starch glycolate, sodium lauryl sulfate, mannitol, lactose, starch, magnesium stearate, polyvinyl pyroUidone, sodium saccharine, cellulose and magnesium carbonate in the formation of a capsule or tablet.
In embodiments involving a capsule, the capsule may be comprise a slow-dissolving polymers. Non-limiting polymers include sodium carboxymethylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose and hydroxyethylcellulose. Other preferred cellulose ethers are known to the skilled person (Alderman, Int. J. Pharm. Tech. & Prod. Mfr., 1984, 5(3): 1-9). Moreover, the polysaccharide material can be directly encapsulated within a capsule or formed into microspheres that are encapsulated. The formation of microspheres may be by a variety of methods known to the skilled person. As a non-limiting example, the polysaccharide(s) are dispersed in a liquid form, such as in an aqueous solution. The liquid is sprayed onto a core particle, such as a nonpareil composed of sugar and/or starch. This forms a microsphere, which may then be dried, or otherwise processed, before being packaged into capsules.
In embodiments involving a tablet, the polysaccharide material can be formed into a solid tablet, optionally with one or more of the excipients listed above. A tablet may be coated by methods known to the skilled person. Solid oral administration can be formulated to give controlled release of the polysaccharide material.
Polysaccharide material may also be formulated into capsule form as a liquid. The liquid may be any suitably formulated for inclusion in a capsule as known to the skilled person. In some embodiments, the liquid is suitably viscous and does not solvate the capsule to result in leakage from the capsule. The liquid may be a preparation that is a variation of those used in other oral administration, such as those in the form of solutions, syrups, or suspensions, all of which may also be used in the practice of the invention. Such liquid preparations can be prepared by conventional means known to the skilled person with pharmaceutically acceptable additives such as, but not limited to, suspending agents, e.g., sorbitol syrup, cellulose derivatives, or hydrogenated edible fats; emulsifying agents, e.g., lecithin or acacia; non-aqueous vehicles, e.g., almond oil, oily esters, ethyl alcohol, or fractionated vegetable oils; and preservatives, e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid. The preparations can also contain buffer salts, flavoring, coloring, and/or sweetening agents as appropriate.
Alternatively, polysaccharide material can be formulated as a food additive. For example, dried polysaccharide can be resuspended in a food substance such as a salad dressing or another sauce or condiment. Alternatively, the material can be formulated into a processed food item. Non-limiting examples include dried foods, canned foods, bars, and frozen foods. Dried foods include dehydrated foods (which are normally rehydrated before consumption), dry cereals, and crackers as non-limiting examples.
In some embodiments, the polysaccharide material can be formulated into a liquid preparation and for administration as a beverage. Such beverage can be alcoholic, nonalcoholic beverage, carbonated, or a health beverage. Such beverage may comprise one or more of the polysaccharides and/or homogenates described herein as well as, optionally, any one or more of the following: a vitamin, electrolyte substitute, caffeine, an amino acid, minerals, artificial and/or natural sweeteners, milk or dry-milk powder, plant phytosterols, and other additives and preserving agents.
Additional carriers of the invention include but are not limited to water, salt solutions (e.g., NaCl), saline, buffered saline, alcohols, glycerol, ethanol, gum arabic, vegetable oils, benzyl alcohols, polyethylene glycols, gelatin, carbohydrates such as lactose, amylose or starch, dextrose, magnesium stearate, talc, silicic acid, viscous paraffin, perfume oil, fatty acid esters, hydroxymethylcellulose, polyvinyl pyrolidone, etc., as well as any two or more of the foregoing in combination.
In some embodiments, the solid or liquid compositions described herein may be advantageously used as a cholesterol lowering composition. Such a composition may comprise 1) a purified microalgal exopolysaccharide or amicroalgal cell homogenate (ie: polysaccharide material) and 2) a carrier suitable for human oral consumption as described. The exopolysaccharide or cell homogenate may be produced from cells of the genus Porphyridium as a non-limiting example. As disclosed herein, the exopolysaccharide may be substantially free of protein.
As described herein, embodiments of the invention include a composition comprising a homogenate of red microalgae cells, such as cells of the genus Porphyridium, and at least one additional cholesterol lowering agent or compound. In some embodiments, the agent or compound is selected from a plant phytosterol, a limonoid, a flavonoid, a tocotrienol, or a combination thereof. In additional embodiments, the agent is an inhibitor of HMG CoA reductase, such as a statin.
In embodiments of a composition comprising red microalgae cell material, such as a homogenate, and a plant phytosterol, the weight/weight ratio of cell material (such as homogenate) to plant phytosterol may range from at least about 0.5:1 to at least about 100:1. Additional non-limiting examples include weight/weight ratios of cell material (such as homogenate) to plant phytosterol of at least about 10:1, at least about 20:1, at least about 30:1, at least about 40:1, at least about 50:1, at least about 60:1, at least about 70:1, at least about 80:1, or at least about 90:1.
Phytosterols are steroids derived from a plant, yeast or fungus. Phytosterols have a hydroxyl group at C-3 and no other functional groups. Plant and animal sterols differ. The term phytosterol and/or phytostanol according to the invention, includes analogues of phytosterols, such as those with a double bond at the 5-position in the cyclic unit as in most natural phytosterols, or one or more double bonds at other positions (e.g. 6, 7, 8(9), 8(14), 14, 5/7, or no double bonds in the cyclic unit as in the stands, or even additional methyl groups as e.g. in alpha- 1 -sitosterol. The invention also includes the use of a mixture of phytosterol and phytostanol with red microalgae cell material.
Representative examples of a phytosterol and/or phytostanols are those obtained from vegetable oil or wood pulp. Non-limiting examples include alpha-sitosterol, beta- sitosterol, gamma-sitosterol, stigmasterol, ergosterol, campesterol, avenasterol, desmosterol, chalinosterol, poriferasterol, clionasterol, sitostanol, alpha-sitostanol, beta-sitostanol, stigmastanol, campestanol or brassicasterol. In addition to these, at least 44 phytosterols have been identified and they may be used in the practice of the invention as deemed appropriate and/or desirable by the skilled person. Additionally, oryzanol may be used. Phytosterols have been reported and are known in the field (see "Advance in Lipid Research", pages 193-218, Paoletti, and Kiritchevsky, (Eds) Academic press, NY, (1993); "Effect of Plant Sterols on Lipids and Atherosclerosis", Pollack, O. J., Pharmac, Ther., 31, 177-208 (1985); Annu Rev Plant Biol. 2004;55:429-57; Metabolism. 2006 Mar;55(3):391 - 5; Am J ClinNutr. 2005 Jun;81 (6): 1351-8; Lipids. 2005 Feb;40(2): 169-74; Lipids. 2003 Apr;38(4):367-75; "Efficacy and safety of plant stanols and sterols in the management of blood cholesterol levels." 2003 Mayo Clin Proc. Aug;78(8):965-78; N Engl J Med. 1995 Nov 16;333(20):1308-12; and U.S. Patents 6,762,312; 6,979,743; 6,929,816; 6,713,118; and 6,383,514, all of which are hereby incorporated by reference as if fully set forth). In optional embodiments, the plant phytosterol is a preparation of wood-derived phytosterol. In some embodiments, the wood-derived phytosterol preparation comprises at least about 8% stanol, at least about 40% sitosterol, and at least about 20% campesterol. In other embodiments, the wood-derived phytosterol preparation comprises between about 5% and about 20% stanol, between about 40% and about 60% sitosterol, and between about 15% and about 35% campesterol.
In embodiments of a composition comprising red microalgae cell material (such as a homogenate) and a limonoid, the weight/weight ratio of cell material (such as homogenate) to limonoid may range from at least about 25:1 to at least about 900:1. Additional non- limiting examples include weight/weight ratios of cell material (such as homogenate) to limonoid of at least about 50:1, at least about 100:1, at least about 200:1, at least about 300:1, at least about 400:1, at least about 500:1, at least about 600:1, at least about 700:1, or at least about 800:1. Limonoid compounds are known to the skilled person, and they include limonin or nomilin, which may be present in a composition as described herein.
In embodiments of a composition comprising red microalgae cell material (such as a homogenate) and a flavonoid, the weight/weight ratio of cell material (such as homogenate) to flavonoid may range from at least about 40:1 to at least about 800:1. Additional non- limiting examples include weight/weight ratios of cell material (such as homogenate) to flavonoid of at least about 70:1, at least about 100:1, at least about 200:1, at least about 300:1, at least about 400:1, at least about 500:1, at least about 600:1, or at least about 700:1.
Flavonoid compounds are known to the skilled person, and they include polymefhoxylated flavones (pmfs), which decrease apoprotein b (a structural protein needed for endogenous synthesis of LDL and cholesterol). The pmfs tangeretin and nobiletin decrease diacylglycerol acetyl transferase (a liver enzyme needed for endogenous synthesis of triglycerides) activity. Some compositions of the invention comprise a flavonoid selected from hesperidin, naringin, naringenin, hesperitin, nobiletin or tangeretin.
In embodiments of a composition comprising red microalgae cell material (such as a homogenate) and a tocotrienol, the weight/weight ratio of cell material (such as homogenate) to flavonoid may range from at least about 30:1 to at least about 900:1. Additional non-limiting examples include weight/weight ratios of cell material (such as homogenate) to flavonoid of at least about 50:1, at least about 100:1, at least about 200:1, at least about 300:1, at least about 400:1, at least about 500:1, at least about 600:1, at least about 700:1, or at least about 800:1. Tocotrienol compounds are known to the skilled person, and some have been reported to inhibit HMG CoA reductase, a liver enzyme responsible for endogenous synthesis of cholesterol. A composition of the invention may comprise one or more tocotrienol, like alpha-tocotrienol, gamma tocotrienol, and delta-tocotrienol as non-limiting examples.
Additional embodiments of the invention include a composition comprising a homogenate of red microalgae cells and at least two, or at least three, compounds selected from a plant phytosterol, a limonoid, a flavonoid, and a tocotrienol. In some embodiments, the composition comprises a plant phytosterol and at least one flavonoid selected from hesperidin, naringin, naringenin, hesperitin, nobiletin and tangeretin.
Descriptions of additional limonoid, flavonoid and tocotrienol compounds for inclusion in a composition of the invention or use in a disclosed method are found in U.S. Patents 6,239,114; 6,251,400; 6,239,114 and 6,987,125; U.S. Patent Application Nos. 09/481,724; 08/938,640; 11/176,593; and 60/560,284; Japanese Patent application PCT/1B98/01721; PCT Patent Application Nos. PCT/lBOl/00256; WO02055071; and PCT/USOl/08395; Canadian Patent Application No. 304202; and European Patent Application Nos. 98947740.1 and 1920415.5, all of which are hereby incorporated by reference as if fully set forth.
Additional embodiments of the invention include methods of formulating the above described compositions. The method may comprise combining or mixing the homogenate and the one or more cholesterol lowering agent or compound. The homogenate may be prepared as described herein, such as by culturing cells of the genus Porphyridium; separating the cells from culture media to prepare a cell material (biomass in this instance); drying (at least partially) the cell material; and homogenizing/disrupting the cell material. Alternatively, the cells and polysaccharides in the media are isolated together and then dried or concentrated (at least partially) before homogenization.
Additional alternative embodiments of the invention include a kit or composition comprising red microalgae cell material, such as cell material of the genus Porphyridium, and at least one cholesterol lowering component. The combination of cell material and the at least one component is formulated, or packaged, for use or sale as a single unit. In some embodiments, the cell material comprises cells per se, while in other embodiments, the cell material is a polysaccharide containing fraction of a preparation of cells (biomass). The cholesterol lowering component may be an agent or compound selected from a plant phytosterol, a limonoid, a flavonoid, a tocotrienol, or a combination thereof as described herein. In additional embodiments, the agent is an inhibitor of HMG CoA reductase, such as a statin.
In some cases, the composition is formulated to comprise the cell material and the at least one component in contact with each other.
In other cases, the cell material is not in contact with the component as these materials are present together in the form of a kit. In the case of a kit, it may further comprise instructions or a label directing the mixing of the cell material (such as a homogenate) and at least one component in a specific ratio, such as by weight or by mole. Alternatively, the instructions or a label may direct ingestion of the cell material and the at least one component in specific dosages. An additional embodiment of the invention includes instructions or a label directing administration of the cell material and at least one component to lower cholesterol levels in a subject or as part of ongoing treatments or therapies to lower cholesterol levels. A further embodiment of the invention includes instructions or a label directing administration of the cell material and at least one component in a specific order, such as ingestion of the cell material and at least one component over a specified time period or with separation by a specific time period.
Further embodiments of the invention include methods of formulating the above described composition comprising cell material and at least one cholesterol lowering agent or compound. The method may comprise combining or mixing cells, optionally dried (at least partially), and the one or more agent or compound. In some embodiments, the method comprises culturing cells of the genus Porphyridium; separating the cells from culture media to prepare a cell containing material (biomass); drying (at least partially) the cell containing material; and mixing the dried material with at least one cholesterol lowering agent or compound. Non-limiting examples of drying methods include tray drying, spin drying, rotary drying, spin flash drying, and lyophilization.
Optionally, the cell containing material is homogenized before mixing with the at least one agent or compound to generate a mixture. Non-limiting examples of methods for homogenization include pressure disruption, sonication, jet milling and ball milling. In some cases, the cell containing material is homogenized such that the homogenate contains at least two times the amount of solvent-available polysaccharide present in a quantity of unhomogenized cells needed to generate the homogenate. In other cases, the cells are washed before homogenization.
Additional embodiments include methods wherein the cells are cultured in at least 100, at least 200, at least 300, at least 400, at least 500, at least 600, or at least 700 mM sulfate. The sulfate may be provided in at least one form selected from Na2SO4-IO H2O, MgSO4*7H20, MnSO4, and CuSO4. Where such cells are homogenized before use, the resultant homogenate may contain sulfated exopolysaccharides with at least about 4.6% sulfur by weight of purified polysaccharide, at least about A.I '5% sulfur by weight of purified polysaccharide, at least about 5.0% sulfur by weight of purified polysaccharide, at least about 5.25% sulfur by weight of purified polysaccharide, at least about 5.5% sulfur by weight of purified polysaccharide, at least about 5.75% sulfur by weight of purified polysaccharide, at least about 6.0% sulfur by weight of purified polysaccharide, at least about 6.25% sulfur by weight of purified polysaccharide, at least about 6.5% sulfur by weight of purified polysaccharide, at least about 6.75% sulfur by weight of purified polysaccharide, or at least about 7.0% sulfur by weight by weight of purified polysaccharide.
The method of preparing the composition may further comprise adding a carrier suitable for human consumption, or a carrier for oral consumption or administration to a mammal, into the mixture of cell material and at least one cholesterol lowering agent or compound.
C. Administration and Methods of Lowering Cholesterol
The cholesterol lowering compositions of the invention may be administered to a subject in need thereof by any appropriate means. Subjects in need of lower cholesterol levels include human beings, who may be tested for serum or plasma cholesterol levels as commonly practiced in clinical medicine by the skilled person. Based on such tests, an elevated cholesterol level in need of lowering may be identified and treated by the methods of the invention. In some embodiments, the cholesterol to be lowered is that of low density lipoprotein (LDL) in serum. In other embodiments, the cholesterol to be lowered is that of Lp(a), a genetic variation of plasma LDL .
The invention includes a method of lowering cholesterol, said method comprising administering a polysaccharide, as disclosed herein, produced by microalgae. In some embodiments, the administering is oral, optionally with a biologically acceptable carrier.
In some embodiments, the polysaccharide is produced by microalgae selected from Table 1. In some embodiments, the polysaccharide is produced by microalgae of the genus Porphyridium. The administered polysaccharide may be a component of a food composition as a non-limiting example. In one range of embodiments, the amount of polysaccharide administered to a human is from about 0.1 to about 50 grams per day. Additional ranges of the invention include an amount of polysaccharide from about 0.25 to about 50 grams per day, about 0.5 to about 5 grams per day, about 0.75 to about 4 grams per day, or about 1 to about 3 grams per day.
Alternatively, the polysaccharide is administered via a composition comprising a homogenate of red microalgae cells as described herein. The amount of the composition may be that which contains sufficient homogenate to provide the equivalent amount of polysaccharide as described above.
Other embodiments of the invention include a method of lowering cholesterol levels in a subject, such as a human patient, by administering a composition comprising a homogenate of red microalgae cells as described herein. In some cases, the method may comprise oral administration of the composition. The amount of the composition to administer, and be sufficient (or effective) to lower serum cholesterol levels, may be determined by the skilled person by routine and/or repetitive methods. In some embodiments, the amount of homogenate administered is in the range of about 0.005 to about 5 grams per kilogram of mammalian body weight per day. Additional non-limiting examples of amounts to use include about 0.01, about 0.025, about 0.05, about 0.075, about 0.1, about 0.25, about 0.5, about 0.75, about 1, about 1.5, about 2, about 2.5, about 3, about 3.5, about 4, or about 4.5 grams per kg of body weight per day.
In some embodiments, the composition comprises at least one cholesterol lowering agent or compound, such as, but not limited to, a plant phytosterol, a limonoid, a flavonoid, a tocotrienol, or a combination thereof. In embodiments comprising a limonoid, the administered amount thereof may be in the range of about 1 to about 500 mg/day, such as about 10, about 20, about 50, about 100, about 150, about 200, about 250, about 300, about 350, about 400, or about 450 mg/day.
In embodiments comprising a flavonoid, the administered amount thereof may be in the range of about 200 to about 5000 mg/day, such as about 250, about 300, about 350, about 400, about 450, about 500, about 600, about 700, about 800, about 900, about 1000, about 1500, about 2000, about 2500, about 3000, about 3500, about 4000, or about 4500 mg/day. In embodiments comprising a tocotrienol, the administered amount thereof may be in the range of about 1 to about 1200 mg/day, such as about 10, about 20, about 50, about 100, about 150, about 200, about 250, about 300, about 350, about 400, about 450, about 500, about 600, about 700, about 800, about 900, about 1000, or about 1100 mg/day.
IV Cosmeceutical Compositions and Topical Application A. General
Compositions, comprising polysaccharides, whole cell extracts, or mixtures of polysaccharides and whole cell extracts, are provided for topical application or non- systemic administration. The polysaccharide may be any one or more of the microalgal polysaccharides disclosed herein, including those produced by a species, or a combination of two or more species, in Table 1. Similarly, a whole cell extract may be that prepared from a microalgal species, or a combination of two or more species, in Table 1. In some embodiments, polysaccharides, such as exopolysaccharides, and cell extracts from microalgae of the genus Porphyridiwn are used in the practice of the invention. A composition of the invention may comprise from between about 0.001% and about 100%, about 0.01% and about 90%, about 0.1% and about 80%, about 1% and about 70%, about 2% and about 60%, about 4% and about 50%, about 6% and about 40%, about 7% and about 30%, about 8% and about 20%, or about 10% polysaccharide, cell extract, by weight.
Topical compositions are usually formulated with a carrier, such as in an ointment or a cream, and may optionally include a fragrance. One non-limiting class of topical compositions is that of cosmeceuticals. Other non-limiting examples of topical formulations include gels, solutions, impregnated bandages, liposomes, or biodegradable microcapsules as well as lotions, sprays, aerosols, suspensions, dusting powder, impregnated bandages and dressings, biodegradable polymers, and artificial skin. Another non-limiting example of a topical formulation is that of an ophthalmic preparation. Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water. Alternatively, the composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2- octyldodecanol, benzyl alcohol and water.
In some embodiments, the polysaccharides contain fucose moieties. In other embodiments, the polysaccharides are sulfated, such as exopolysaccharides from microalgae of the genus Porphyridium. In some embodiments, the polysaccharides will be those from a Porphyridium species, such as one that has been subject to genetic and/or nutritional manipulation to produce polysaccharides with altered monosaccharide content and/or altered sulfation. In additional embodiments, a composition of the invention comprises a microalgal cell homogenate and a topical carrier. In some embodiments, the homogenate may be that of a species listed in Table 1 or may be material produced by a species in the table.
In further embodiments, a composition comprising purified microalgal polysaccharide and a carrier suitable for topical administration also contains a fusion (or chimeric) protein associated with the polysaccharide. In some embodiments, the fusion protein comprises a first protein, or polypeptide region, with at least about 60% amino acid identity with the protein of SEQ ID NO: 28. In other embodiments, the first protein has at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 98%, or higher, amino acid identity with the sequence of SEQ ID NO:28.
The fusion protein may comprise a second protein, or polypeptide region, with a homogenous or heterologous sequence. Non-limiting examples of the second protein include an antibody and an enzyme. In optional embodiments, the enzyme is superoxide dismutase, such as that has at least about 60% amino acid identity with the sequence of SEQ ID NO: 14, SEQ ID NO: 15, and a superoxide dismutase selected from Table 16 as non- limiting examples. In some embodiments, the superoxide dismutase has at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 98%, or higher, amino acid identity with the sequences of SEQ ID NO: 14 or 15 or from Table 16.
In other embodiments, the second protein is an antibody. Non-limiting examples of antibodies for use in this aspect of the invention include an antibody that selectively binds to an antigen from a pathogen selected from HIV, Herpes Simplex Virus, gonorrhea, Chlamydia, Human Papillomavirus, and Trichomoniasis. In some embodiments, the antibody is a humanized antibody.
B. Methods of Formulation
Polysaccharide compositions for topical application can be formulated by first preparing a purified preparation of polysaccharide. As a non-limiting example, the polysaccharide from aqueous growth media is precipitated with an alcohol, resuspended in a dilute buffer, and mixed with a carrier suitable for application to human skin or mucosal tissue, including the vaginal canal. Alternatively, the polysaccharide can be purified from growth media and concentrated by tangential flow filtration or other filtration methods, and formulated as described above. Intracellular polysaccharides can be also formulated in a similar or identical manner after purification from other cellular components.
As a non-limiting example, the invention includes a method of formulating a cosmeceutical composition, said method comprising culturing microalgal cells in suspension under conditions to allow cell division; separating the microalgal cells from culture media, wherein the culture media contains exopolysacchari.de molecules produced by the microalgal cells; separating the exopolysaccharide molecules from other molecules present in the culture media; homogenizing the microalgal cells; and adding the separated exopolysaccharide molecules to the cells before, during, or after homogenization. In some embodiments, the microalgal cells are from the genus Porphyridium.
Examples of polysaccharides, both secreted and intracellular, that are suitable for formulation with a carrier for topical application are listed in Table 1.
Examples of carriers suitable for formulating polysaccharide are described above. Ratios of homogenatercarrier are typically in the range of about 0.001 : 1 to about 1:1 (volume:volume), although the invention comprises ratios outside of this range, such as, but not limited to, about 0.01:1 and about 0.1:1.
Microalgal cellular extracts can also be formulated for topical administration. It is preferable but not necessary that the cells are physically or chemically disrupted as part of the formulation process. For example, cells can be centrifuged from culture, washed with a buffer such as 1.0 mM phosphate buffered saline, pH 7.4, and sonicated. Preferably the cells are sonicated until the cell walls have been substantially disrupted, as can be determined under a microscope. For example, Porphyridium sp. cells can be sonicated using a Misonix sonicator as described in Example 15.
Cells can also be dried and ground using means such as mortar and pestle, colloid milling, ball milling, or other physical method of breaking cell walls.
After cell disruption, cell homogenate can be formulated with carrier and fragrance as described above for polysaccharides.
C. Co-Administered Compositions
Topical compositions can comprise a portion of a complete composition sold as a single unit. Other portions of the complete compositions can comprise an oral supplement intended for administration as part of a regime for altering skin appearance. Because the top layers of the skin contain dead cells, nutrients delivered via capillaries cannot reach the outer layers of cells. The outer layers of cells must be provided with nutrients though topical administration. However, topical administration is not always an effective method of providing nutrients to deep layers of skin that contain living cells. The compositions provided herein comprise both topical compositions that contain algal polysaccharides and/or cellular extracts as well as oral compositions comprising nutraceutical molecules such as purified polysaccharides, whole cell extracts, carotenoids, polyunsaturated fatty acids, and other molecules that are delivered to the skin via capillaries. The combined effect of the topical and oral administration of these molecules and extracts provides a benefit to skin health that is additive or synergistic compared to the use of only a topical or only an orally delivered product.
Examples of the topical components of the composition include exopolysaccharide from Porphyridium cruentum, Porphyridium sp., list others. Other components of the topical composition can include polysaccharides and/or cell extracts from species listed in Table 1.
Cellular extracts for topical administration can also include cellular homogenates from microalgae that have been genetically engineered. For example, homogenates of Porphyridium sp. that have been engineered to express an exogenous gene encoding superoxide dismutase can be formulated for topical administration. Other genes that can be expressed include carotenoid biosynthesis enzymes and polyunsaturated fatty acid biosynthesis enzymes.
Examples of compositions for oral administration include one or more of the following: DHA, EPA, ARA, lineoileic acid, lutein, lycopene, beta carotene, braunixanthin, zeaxanthin, astaxanthin, linoleic acid, alpha carotene, vitamin C and superoxide dismutase. Compositions for oral administration usually include a carrier such as those described above. Oral compositions can be formulated in tablet or capsule form. Oral compositions can also be formulated in an ingestible form such as a food, tea, liquid, etc.
In a preferred embodiment, at the topical composition and the oral composition both contain at least one molecule in common. For example, the topical composition contains homogenate of Porphyridium cells that contain zeaxanthin, and the oral composition contains zeaxanthin. In another embodiment, the topical composition contains homogenate of Porphyridium cells that contain polysaccharide, and the oral composition contains polysaccharide purified from Porphyridium culture media.
The compositions described herein are packaged for sale as a single unit. For example, a unit for sale comprises a first container holding a composition for topical administration, a second container holding individual doses of a composition for oral administration, and optionally, directions for co-administration of the topical and oral composition.
Some embodiments of the invention include a combination product comprising 1) a first topical composition comprising a microalgal extract or other composition and a carrier suitable for topical application to skin; and 2) a second composition comprising at least one compound in common with the first composition and optionally a carrier suitable for human oral consumption; wherein the first and second compositions are packaged for sale as a single unit. Thus the invention includes co-packaging of the two compositions, optionally with a instructions and/or a label indicating the identity of the contents and/or their proper use.
In some embodiments the first topical composition is selected from an algae extract, optionally from a species selected from Table 1 , a vitamin C preparation (such as L-ascorbic acid or ascorbyl palmitate), and a vitamin A preparaion (such as d-alpha-tocopherol). In some embodiments the second oral compositions is selcted from an algae extract, optionally from a species selected from Table 1, a vitamin C composition (such as L-ascorbic acid or ascorbyl palmitate), a vitamin A preparaion (such as d-alpha-tocopherol), a polyunsaturated fatty acid (such as Eicosapentaenoic Acid (EPA) or Docosahexaenoic Acid (DHA), zeaxanthm, lutein, beta carotene, and a coenzyme Q preparation (such as ubiquinol-10). Prefferred algae extracts contain at least a subset of vitamin C, vitamin A, polyunsaturated fatty acids such as EPA or DHA, and carotenoids such as zeaxanthin, lutein, beta carotene, and cofactors such as coenzyme Q.
D. Methods of Cosmetic Enhancement
In a further aspect, the invention includes a polysaccharide composition suitable for injection into skin to improve its appearance. In some embodiments, the injection is made to alleviate or eliminate wrinkles. In other embodiments, the treatment reduces the visible signs of aging and/or wrinkles.
As known to the skilled person, human skin, as it ages, gradually loses skin components that keep skin pliant and youthful-looking. The skin components include collagen, elastin, and hyaluronic acid, which have been the subject of interest and use to improve the appearance of aging skin.
The invention includes compositions of microalgal polysaccharides, microalgal cell extracts, and microalgal cell homogenates for use in the same manner as collagen and hyaluronic acid. In some embodiments, the polysaccharides will be those of from a Porphyridium species, such as one that has been subject to genetic and/or nutritional manipulation to produce polysaccharides with altered monosaccharide content and/or altered sulfation. In some embodiments, the polysaccharides are formulated as a fluid, optionally elastic and/or viscous, suitable for injection. The compositions may be used as injectable dermal fillers as one non-limiting example. The injections may be made into skin to fill-out facial lines and wrinkles. In other embodiments, the injections may be used for lip enhancement. These applications of polysaccharides are non-limiting examples of non- pharmacological therapeutic methods of the invention.
In further embodiments, the microalgal polysaccharides, cell extracts, and cell homogenates of the invention may be co-formulated with collagen and/or hyaluronic acid (such as the Restylane® and Hylaform® products) and injected into facial tissue. Non- limiting examples of such tissue include under the skin in areas of wrinkles and the lips. In a preferred embodiment, the polysaccharide is substantially free of protein. The injections may be repeated as deemed appropriate by the skilled practitioner, such as with a periodicity of about three, about four, about six, about nine, or about twelve months. In another preferred embodiment, a hyaluronic acid material is mixed with a polysaccharide from the genus Porphyridium prior to co-administration. The invention in this particular embodiment provides longer half-life to the hyaluronic acid due to the potent inhibition of hyaluronidase by polysaccharides isolated from microalgae from the genus Porphyridium. This allows for less injections to a patent.
Thus the invention includes a method of cosmetic enhancement comprising injecting a polysaccharide produced by microalgae into mammalian skin. The injection may be of an effective amount to produce a cosmetic improvement, such as decreased wrinkling or decreased appearance of wrinkles as non-limiting examples. Alternatively, the injection may be of an amount which produces relief in combination with a series of additional injections. In some methods, the polysaccharide is produced by a microalgal species, or two or more species, listed in Table 1. In one non-limiting example, the microalgal species is of the genus Porphyridium and the polysaccharide is substantially free of protein.
V Compositions for Non-Systemic Administration of Polysaccharides
A. General
Compositions for non-systemic administration include those formulated for localized administration with little or slow release to other parts of a treated subject's body. Non-limiting examples of non-systemic administration include intravaginal application such as via a suppository, cream or foam; injection into a joint between bones; intravitreous or intraocular administration; and rectal administration via suppository, irrigation or other suitable means. In some embodiments, the composition is formulated for the treatment of sexually transmitted diseases, such as those caused by viral agents.
Polysaccharides from microalgae provided herein posses potent antiviral activity (see references cited in Table 1). In additional embodiments, polysaccharides with lubricant properties (see for example Porphyridium and Chlorella polysaccharides) are used in the practice of certain aspects of the invention. These polysaccharides maybe formulated in solutions that are added to prophylactic devices. Moreover, the polysaccharides may be one or more described herein, optionally sulfated. In many embodiments, the polysaccharide is produced by a microalgal species, or two or more species, listed in Table 1. In some embodiments, the microalgae is Porphyridium sp. or Porphyridium omentum.
Thus, the invention includes a sexually transmitted disease prevention composition, said composition comprising 1) a solution comprising a polysaccharide produced from microalgae; and 2) a prophylactic device. In some embodiments, the solution and device are kept separate, but packaged together as a single unit for sale. The solution may be applied to the device by the end user before actual use. Alternatively the solution and device are packaged so that the solution is in direct contact with the device. The prophylactic devices include, but are not limited to, condoms, sponges, and diaphragms.
In some embodiments, the devices are packaged with a lubricant. In other embodiments, the polysaccharide acts as a lubricant and so no other lubricant is needed. In such embodiments, the substance in the composition providing a lubricant function and the substance in the composition providing antiviral activity are the same substance. Alternatively, a combination of a lubricant, such as a cream or lotion, with the polysaccharide of the invention may be used.
In some embodiments, the polysaccharide is in a composition with a carrier used with a prophylactic device described above. Non-limiting examples of a carrier include a spermicide and a lubricant. In other embodiments of the invention, a triple composition, comprising spermicide, lubricant and the polysaccharide, may be used.
In further embodiments, the polysaccharide is associated with a fusion (or chimeric) protein comprising a first protein (or polypeptide region) with at least about 60% amino acid identity with the protein of SEQ ID NO: 28. In some cases, the first protein has at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 98%, or higher, amino acid identity with the sequence of SEQ ID NO:28, and further wherein the fusion protein has the ability to specifically bind to the polysaccharide, preferably with high affinity.
The fusion protein may comprise a second protein, or polypeptide region, with a homogenous or heterologous sequence. One non-limiting example of the second protein is an antibody. In some embodiments, the antibody is selective for binding to an antigen of a pathogen, or opportunistic organism, involved in a sexually transmitted disease. Non- limiting examples of antibodies include those that bind an antigen from a pathogen selected from HIV, Herpes Simplex Virus, gonorrhea, Chlamydia, Human Papilloma Virus, and Trichomoniasis .
In other embodiments, the compositions are formulated for improving joint lubrication or treating joint disorders. As described above, microalgal polysaccharides may be used in the same manner as, or in combination with, hyaluronic acid in some compositions of the invention. Hyaluronic acid, or hyaluronan, is used to lubricate joints, such as in viscosupplementation. As a non-limiting example, SYNVISC® (Genzyme Corporation) is an FDA-approved agent which is injected into knee joints to provide lubrication. The elastic and viscous nature of the fluid allows it to function in absorbing shock and improve proper knee movement and flexibility. Other similar hyaluronic acid products include Hylartil®, Arthrease®, Orthovisc® and Artval®. In another preferred embodiment, a hyaluronic acid material is mixed with a polysaccharide from the genus Porphyridium prior to injection into a mammalian joint. The invention in this particular embodiment provides longer half-life to the hyaluronic acid due to the potent inhibition of hyaluronidase by polysaccharides isolated from microalgae from the genus Porphyridium. This allows for less injections to a patent.
Microalgal polysaccharides of the invention are also formulated as fluids with elastic and/or viscous properties such that they may serve as replacements for normal joint fluid. Polysaccharides from the red microalgae Porphyidium sp. have desirable load bearing and shear properties. Polysaccharides with average molecular weights of about 2 to about 7 megadaltons in solution have been found to have very low coefficients of friction (μ <0.01) at low compressions, and increasing only to μ =0.015 at 10 MPa. The low friction, and resistance under high pressure make the polysaccharides highly suitable for biolubrication, such as in human joint lubrication. Advantageously, the polysaccharides are not degraded by hyaluronidase, which degrades hyaluronic acid; are resistant to elevated temperatures; and are anti-inflammatory and anti-irritating. See for example, Golan et al., "Characterization of a Superior Bio-Lubricant Extracted from a Species of Red Microalga " The 39th Annual Meeting of the Israel Society for Microscopy, Ben Gurion University, May 19th, 2005, Poster Abstracts (at www.technion.ac.il/technion/materials/ism/ISM2005_posters_abstracts.html); and Gourdon et al. "Superlubricity of a natural polysaccharide from the alga Porphyridium sp " Abstract Submitted for the March 2005 Meeting of The American Physical Society, Abstract V31.00010 (at absimage.aps.org/image/MWS_MAR05-2004-006269.pdf).
B. Methods of Use
The polysaccharides of the invention may be used in the same or a similar manner, in some embodiments, the polysaccharides will be those from a. Porphyridium species, such as one that has been subject to genetic and/or nutritional manipulation to produce polysaccharides with altered monosaccharide content, m some embodiments, a fluid containing one or more polysaccharides is injected into a joint to. alleviate joint pain, such as, but not limited to, arthritis and osteoarthritis. Non-limiting examples of joint pain include pain of the knee, shoulder, elbow, and wrist joints. Subjects afflicted with, suffering from, or having joint pain may be diagnosed and/or identified by a skilled person in the field using any suitable method. Non-limiting examples include signs of inflammation, like swelling, pain, or redness; excess fluid in the joint; the need for physical therapy; pain during exercise.
In other embodiments, the polysaccharides of the invention, whether used alone or in combination with hyaluronic acid, are used after the failure, or ineffectiveness, of non- drug treatments or drug therapy for joint pain. Non-limiting examples of non-drug treatments that may be ineffective include avoidance of activities that cause the joint pain, exercise, physical therapy, and removal of excess fluid. Non-limiting examples of drug therapy that may be ineffective include pain relievers, such as acetaminophen and narcotics; anti-inflammatory agents, such as aspirin and other nonsteroidal anti-inflammatory drugs (NSAIDs) such as ibuprofen and naproxen; and injection of steroids.
The invention includes a method of mammalian joint lubrication. Mammalian joint lubrication is used to treat conditions such as osteoarthritis, joint trauma, rheumatoid arthritis, and other degenerative conditions affecting the mammalian joint. Mammalian joints include knees, hips, ankles, shoulders, and other joints. The method comprises injecting a microalgal polysaccharide of the invention into a cavity containing synovial fluid. The injection may be of an effective amount to produce relief from one or more symptoms of joint pain or discomfort that is alleviated by joint lubrication. Alternatively, the injection may be of an amount which produces relief in combination with a series of additional injections. In some methods, the polysaccharide is produced by a microalgal species, or two or more species, listed in Table 1. In one non-limiting example, the microalgal species is of the genus Porphyridium.
In further embodiments, the methods may also comprise treatment with one or more of the non-drug treatments or drug therapies described herein. As a non-limiting example, injection of a joint lubricating composition of the invention may be combined with administration of an anti-inflammatory agent and optionally physical therapy.
For systemic administration, polysaccharides can be formulated with carriers, excipients, and other compounds, pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d.alpha-tocopherol polyethyleneglycol 1000 succinate, or other similar polymeric delivery matrices or systems, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat. Cyclodextrins such as alpha-, beta-, and gamma-cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3-hydroxypropyl-beta-cyclodextrins, or other solublized derivatives may also be advantageously used to enhance delivery of therapeutically-effective plant essential oil compounds of the present invention.
The polysaccharide compositions of this invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir, however, oral administration or administration by injection is preferred. The pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles. In some cases, the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form. The term parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
The polysaccharide compositions may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are mannitol, water, Ringers solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant such as Ph. HeIv or a similar alcohol.
Sterile injectable polysaccharide compositions preferably contain less than 1% protein as a function of dry weight of the composition, more preferably less than 0.1% protein, more preferably less than 0.01% protein, less than 0.001% protein, less than 0.0001% protein, more preferably less than 0.00001% protein, more preferably less than 0.000001% protein.
The polysaccharide compositions of the present invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, and aqueous suspensions and solutions. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
The polysaccharide compositions of the present invention may also be administered in the form of suppositories for rectal administration. These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components. Such materials include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
VI Food Additive and Nutraceutical Compositions
A. Food Compositions
The invention further provides for the use of polysaccharides as food additives. In some embodiments, the polysaccharides are used to aid in keeping food products homogenous and/or thicken food products, such as in a manner analogous to various gums (e.g. xanthan gum or gum Arabic) known to the skilled person. In these contexts, the polysaccharides act as a hydrocolloid polysaccharide and may be used in the same manner as other hydrocolloid polysaccharides. See for example U.S. Patent 5,126,158. In other embodiments, the polysaccharides are used to stabilize or emulsify foods. These uses of the polysaccharides are based upon their rheological properties, which are similar or superior to those of previously used gums and additives. Tn some embodiments, the polysaccharides will be those of from a Porphyridium species, such as one that has been subject to genetic and/or nutritional manipulation to produce polysaccharides with altered monosaccharide content and/or altered sulfation.
The methods of the invention thus include a method comprising addition of one or more polysaccharides of the invention to a food product. The addition may be for use as a thickener or an emulsifier, such as to keep foods homogenous. In some embodiments, the addition is to a beverage product, such as to thicken it or improve its texture, appearance, or "feel" upon consumption. In other embodiments, the method stabilizes and/or emulsifies a food composition or product. The method may comprise adding one or more microalgal polysaccharide, as described herein, to the food composition or product. Non-limiting examples include polysaccharides produced by microalgae described herein. In some instances, the microalgae is of the genus Porphyridium.
In some embodiments, a microalgal cell material (such as a homogenate) of the invention may be incorporated into a food, a food supplement, a liquid, a beverage, an emulsion, a powder, or mixtures thereof for ingestion by a mammal. In some cases, the cell material is formulated as a composition for addition to another food substance, such as for sprinkling, or spreading, on a salad, bread, or cereal as non-limiting examples. In further embodiments, the cell material may be incorporated into a cereal product, such as a ready- to-eat variety; incorporated into a baked product, such as a bread, muffin, or waffle; incorporated into a pasta, healthy dessert, snack (such as an athletic bar or healthy drink as non-limiting examples) or a high fiber food.
B. Nutraceuticals
In another aspect, the invention includes nutraceutical compositions comprising one or more polysaccharides, or microalgal cell extract or homogenate, of the invention. A nutraceutical composition serves as a nutritional supplement upon consumption. In other embodiments, a nutraceutical may be bioactive and serve to affect, alter, or regulate a bioactivity of an organism.
A nutraceutical may be in the form of a solid or liquid formulation. In some embodiments, a solid formulation includes a capsule or tablet formulation as described above. In other embodiments, a solid nutraceutical may simply be a dried microalgal extract or homogenate, as well as dried polysaccharides per se. Tn liquid formulations, the invention includes suspensions, as well as aqueous solutions, of polysaccharides, extracts, or homogenates.
The methods of the invention include a method of producing a nutraceutical composition. Such a method may comprise drying a microalgal cell homogenate or cell extract. The homogenate may be produced by disruption of microalgae which has been separated from culture media used to propagate (or culture) the microalgae Thus in one non- limiting example, a method of the invention comprises culturing red microalgae; separating the microalgae from culture media; disrupting the microalgae to produce a homogenate; and drying the homogenate. In similar embodiments, a method of the invention may comprise drying one or more polysaccharides produced by the microalgae.
In some embodiments, a method of the invention comprises drying by tray drying, spin drying, rotary drying, spin flash drying, or lyophilization. In other embodiments, methods of the invention comprise disruption of microalgae by a method selected from pressure disruption, sonication, and ball milling
In additional embodiments, a method of the invention further comprises formulation of the homogenate, extract, or polysaccharides with a carrier suitable for human consumption. As described herein, the formulation may be that of tableting or encapsulation of the homogenate or extract.
In further embodiments, the methods comprise the use of microalgal homogenates, extracts, or polysaccharides wherein the cells contain an exogenous nucleic acid sequence, such as in the case of modified cells described herein. The exogenous sequence may encode a gene product capable of being expressed in the cells or be a sequence which increases expression of one or more endogenous microalgal gene product.
Non-limiting examples of the latter include insertion of regulator regions which increase expression of an endogenous microalgal gene and insertion of additional copies of an endogenous microalgal gene to increase copy number. Thus some embodiments of the invention include microalgal cells expressing an exogenous gene which increases production of a small molecule naturally produced by the microalgae or which induces the microalgae to produce, or directs the production of, a small molecule not naturally produced by the microalgae. In other embodiments, the increased expression of an endogenous microalgal gene or insertion of additional copies of an endogenous microalgal gene to increase copy number is used to increase production of a small molecule normally produced by the microalgae.
In yet further embodiments, the microalgal homogenates, extracts, or polysaccharides are from cells containing a modification to an endogenous nucleic acid sequence. One non-limiting example includes modified microalgal cells wherein an endogenous repressor nucleic acid sequence, or sequence encoding a proteinaceous or RNA gene product, is removed or inhibited such that production of a small molecule normally produced by the microalgae is increased.
Of course the invention includes embodiments wherein nucleic acid modification as described herein increases production of more than one microalgal small molecule.
In some embodiments, the small molecule of a microalgal cell which is increased by these methods of the invention is a carotenoid. Non-limiting examples of carotenoids include lycopene, lutein, beta carotene, zeaxanthin. hi other embodiments, the small molecule is a polyunsaturated fatty acid, such as, but not limited to, EPA, DHA, liholeic acid and ARA.
In additional aspects, the invention includes a nutraceutical composition prepared by a method described herein, hi some embodiments, the composition comprises homogenized red microalgal cells and a carrier suitable for human consumption, hi other embodiments, the carrier is a food product or composition. The microalgal cells may be genetically modified as described above to result in red microalgae which produce an increased amount of a small molecule naturally produced by the red microalgae; or to produce a small molecule not naturally produced by the microalgae. In one non-limiting example, the small molecule is DHA. The invention further provides for a combination composition wherein a microalgal homogenate further comprises an exopolysaccharide produced by the red microalgae. In some embodiments, the exopolysaccharide has been purified from culture media used to grow the red microalgae. The exopolysaccharide may be added to the cells before, during, or after homogenization. In another combination composition, a microalgal homogenate further comprises an exogenously added molecule, such as, but not limited to, EPA, DHA, linoleic acid, ARA5 lycopene, lutein, beta carotene, and zeaxanthin.
A nutraceutical of the invention may also be a composition comprising a purified first polysaccharide produced from a microalgal species listed in Table 1 and a carrier suitable for human consumption. Non-limiting examples of the polysaccharides include sulfated molecules as well as polysaccharides with an average molecular weight (MW) of the polysaccharide is between about 2 and about 7 million Daltons (MDa). In some embodiments, the polysaccharide has an average MW of about 3, about 4.5, about 5, or about 6 MDa. In other embodiments, the average MW is below 2 MDa5 such as below about 1, below about 0.8, below about 0.6, below about 0.4, or below about 0.2 MDa.
In some embodiments, the composition contains between 1 microgram and 50 grams of one type of microalgal polysaccharide. Alternatively, the composition contains more than one type of microalgal polysaccharide, such as one or more additional polysaccharide. In compositions with more than one type of polysaccharide, at least one polysaccharide is optionally from a non-microalgal source, such as a non-microalgal species. In some embodiments, the additional polysaccharide is beta glucan. In further embodiments, a composition further comprises a plant phytosterol.
In some aspects, a composition comprising both a microalgal homogenate and a polysaccharide, such as an exopolysaccharide, is disclosed herein. The composition may comprise homogenized microalgae and isolated or purified or semi-purified exopolysaccharide(s), wherein the composition is a percentage of exopolysaccharide by weight ranging from up to about 1% to up to about 20%, or higher. The remaining portion of the composition may be the homogenate or other carriers and excipients as desired for a composition, nutraceutical, or cosmeceutical of the invention. In some embodiments, the percentage of exopolysaccharide is up to about 2%, up to about 5%, or up to about 10%. This type of combination composition may be prepared by any appropriate means known to the skilled person, including preparing of each component separately and then combining them. In other methods, formulation of a composition comprises subjected a microalgal culture containing exopolysaccharides to tangential flow filtration to concentrate the material and then diafiltration until the composition is substantially free of salts, wherein the cells and exopolysaccharide are both retained in the retentate. The material can also be partially concentrated, diafiltered, and then concentrated further, and this regime can also be used on supernatant free of cells where the exopolysaccharide is retained. The exopolysaccharides may be those produced by the microalgae during culture or may be exogenously added to the culture before processing. The filtered material may then be homogenized or dried as described herein.
Other combination products are including in the invention. In some embodiments, a combination of a first composition for topical application and second composition for consumption is provided. In some embodiments, the first composition may be a topical formulation or non-systemic formulation, optionally a cosmeceutical, as described herein. Preferably, the first composition comprises a carrier suitable for topical application to skin, such as human skin. Non-limiting examples of the second composition include a food composition or nutraceutical as described herein. Preferably, the second composition comprises at least one carrier suitable for human consumption, such as that present in a food product or composition.
In some embodiments, the first and second compositions contain at least one compound in common. Non-limiting examples include one polysaccharide or one carrier in common. In other examples, the at least one compound is selected from DHA, EPA, ARA, lycopene, lutein, beta carotene, zeaxanthin, linoleic acid, vitamin C, and superoxide dismutase.
Combination products of the invention may be packaged separately for subsequent use together by a user or packaged together to facilitate purchase and use by a consumer. Packaging of the first and second compositions may be for sale as a single unit.
C. Methods of Use
A polysaccharide (as well as homogenate or extract) containing food product or nutraceutical of the invention maybe consumed as a source of nutrition and/or sustenance. Thus the invention includes methods of providing food, nutrition or sustenance to a subject, such as a human being, by administration of a composition or nutraceutical as described herein. While a food product may be a primary source of sustenance, a nutraceutical may be used as a nutritional supplement. Thus the invention also includes methods of administering both to a subject. The administered food product may comprise a polysaccharide, extract, or homogenate as described herein. In some embodiments, the polysaccharide, extract or homogenate is used to thicken, stabilize or emulsify foods.
In other aspects, other methods for the use of a polysaccharide containing composition, including those containing a microalgal homogenate or extract of the invention, are disclosed. In some methods, the composition is used to regulate, or aid in the regulation of insulin. Administration of algal polysaccharides included in the invention reduces insulin secretion in response to a given stimulus. Subjects, including human beings, in need of insulin regulation may be identified by any means known to the skilled person. In some embodiments, the subject is identified as being at risk for diabetes by a skilled clinician. Being at risk includes having one or more risk factors, as assessed by the skilled . person, which increase the chances of needing insulin regulation and/or having diabetes. Non-limiting examples of risk factors include those of lifestyle, behavior, health status, disease, and medication use. In some embodiments, the risk factors may amount to the present of "pre-diabetes" or "metabolic disease".
Non-limiting examples of lifestyle factors include inactivity, stress, diet, and aging. Non-limiting examples of behavior factors include levels of sexual activity, smoking, alcohol use, and drug use. Non-limiting examples of health status factors include obesity, cholesterol, diabetes, immunosuppression, and hypertension as well as gender status as a woman, such as pregnancy, childbirth, and menopause. The compositions are particularly useful for lowering cholesterol levels in patients having abnormally high levels of cholesterol of at least 240 mg/dL total cholesterol, at least 160 mg/dL LDL cholesterol, no more than 40 mg/dL HDL cholesterol, and/or at least 400 mg/dL triglycerides.
Non-limiting examples of diseases include HIV, heart, cancer, and autoimmune diseases. Non-limiting examples of medications include use of contraceptives and steroids.
A nutraceutical of the invention may be administered to a subject found to have one or more of these risk factors sufficient to warrant conservative or aggressive treatment of the subject. The determination or diagnosis of risk factor presence may be conducted by a skilled person, such as a clinician. Non-limiting examples of conservative treatment methods may comprise administration of a polysaccharide composition of the invention optionally in combination of one or more alterations in activity to reduce one or more risk factors. Alternatively, the methods may be in the absence of other treatment for insulin malfunction or misregulation, pre-diabetes, or metabolic disease.
Non-limiting examples of aggressive treatment include active administration of a bioactive agent to a subject afflicted with diabetes or insulin misregulation or malfunction. Administration of a bioactive agent includes insulin injection to maintain glucose levels in a subject.
In some embodiments, a method of regulating insulin is provided. Such a method may comprise administering a polysaccharide produced by microalgae as described herein. The polysaccharides may reduce the need for other agents, such as a bioactive agent, that regulate insulin.
In further aspects, antioxidant properties of microalgal polysaccharides may be utilized to treat subjects in need of antioxidant activity. Polysaccharides with antioxidant activity may be identified by.suitable means known to the skilled person. In some embodiments, the polysaccharides will be those from a Porphyridium species, such as one that has been subject to genetic and/or nutritional manipulation to produce polysaccharides with altered monosaccharide content and/or altered sulfation.
In some embodiments, antioxidant polysaccharides are used to inhibit, reduce or treat undesired inflammation. The inflammation can be the result of several diseases including autoimmune diseases, graft versus host disease, host versus graft disease, or pathogenic infections. In some embodiments, the polysaccharides will be those from a Porphyridium species, such as one that has been subject to genetic and/or nutritional manipulation to produce polysaccharides with altered monosaccharide content and/or altered sulfation.
Therefore, aspects of the invention include a method of reducing reactive oxygen species (ROS) in a mammal. In some embodiments, the method is used to prevent or treat a disease or unwanted condition associated with ROS or oxidative stress. Non-limiting examples of such a disease or unwanted condition include a neurodegenerative condition, atherosclerosis, metabolic syndrome, erectile dysfunction, transplant rejection, cancer, cardiovascular disease, hypertension, ischemia, reperfusion injury, rheumatoid arthritis, and aging, which are all conditions that are associated with increased in reactive oxygen species.
In some embodiments, a method comprises administering a composition comprising red microalgae cell material (such as a homogenate) as described herein. In other embodiments, the composition comprises one or more other agents or compounds with antioxidant activity. In further embodiments, the method may be used to lower the level of ROS, or reduce or decrease the amount of damage caused by ROS. The amount of the composition may be any that is effective or sufficient to produce a desired improvement or therapeutic benefit. The method may be used to prevent, treat, or alleviate a symptom of, a neurodegenerative disease or disorder. Non-limiting examples of a neurodegenerative disease or disorder include dementias (e.g., senile dementia, memory disturbances/memory loss, dementias caused by neurodegenerative disorders (e.g., Alzheimer's, Parkinson's disease, Parkinson's disorders, Huntington's disease (Huntington's Chorea), Freidreich's ataxia, Lou Gehrig's disease, multiple sclerosis, Pick's disease, Parkinsonism dementia syndrome), progressive subcortical gliosis, progressive supranuclear palsy, thalmic degeneration syndrome, hereditary aphasia, amyotrophic lateral sclerosis, Shy-Drager syndrome, and Lewy body disease; vascular conditions (e.g., infarcts, hemorrhage, cardiac disorders); mixed vascular and Alzheimer's; bacterial meningitis; Creutzfeld-Jacob Disease; and Cushing's disease. In alternative embodiments, the method may be used to delay the onset of one or more of the above diseases or disorders in a mammal.
The method may also be used to prevent, treat, or alleviate metabolic syndrome in a human subject, which is characterized by at least three of the following: enlargement of the waist diameter, a higher level of arterial pressure, a higher level of low density lipoprotein cholesterol, a higher level of glycemia, and reduction of high density lipoprotein cholesterol (see for example, Palomo et al. Int. J. MoL Med. (2006) 18(5):969-74).
The method may also be used to prevent, treat, or alleviate a symptom of, erectile dysfunction. The involvement of ROS in the disorder has been reported by Jeremy et al. (Int. J. Impot. Res. 2006 Oct 19; Epub ahead of print).
The method may also be used to prevent, treat, or alleviate a symptom of, transplant rejection, such as in the case of organ or tissue transplant as non-limiting examples. The involvement of ROS in transplant rejection is known to the skilled person. See for example Cell Immunol. 2006 Jun;241(2): 59-65.
The method may also be used to prevent, treat, or alleviate a symptom of aging. The involvement of ROS in aging has been reported by Valko et al. (Int. J. Biochem Cell Biol. (2007) 39(l):44-84 Epub 2006 Aug 4).
The invention includes a method to treat inflammation. Such a method may comprise administering a polysaccharide containing composition of the invention to a subject in need of anti-inflammatory activity. The polysaccharide may be one or more produced by microalgae described herein. The administering may be by a variety of means, including direct transfer to a tissue or subject via an intramuscular, intradermal, subdermal, subcutaneous, oral, parenteral, intraperitoneal, intrathecal, or intravenous procedure. Alternatively, a scaffold or binding protein can be placed within a cavity of the body, such as during surgery, or by inhalation, or vaginal or rectal administration.
In prophylactic applications, pharmaceutical compositions or medicaments are administered to a patient susceptible to, or otherwise at risk of, a disease or condition, such as excess cholesterol, inflammation, low insulin, inadequate joint lubrication in an amount sufficient to eliminate or reduce the risk, lessen the severity, or delay the outset of the disease, including biochemical, histologic and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease. In therapeutic applications, compositions or medicants are administered to a patient suspected of, or already suffering from such a disease in an amount sufficient to cure, or at least partially arrest, the symptoms of the disease (biochemical, histologic and/or behavioral), including its complications and intermediate pathological phenotypes in development of the disease.
A microalgal cell material containing composition of the invention may also be used in an effective or sufficient amount to produce a desired improvement or therapeutic benefit in decreasing mammalian body fat (such as via a hypolipidemic effect), controlling or preventing weight gain in a mammal, reducing the body weight in a mammal, increasing satiety (or decreasing appetite) in a mammal, increasing energy expenditure (or energy use) in a mammal, reducing inflammation in a mammal, or preventing and/or treating atherosclerosis in a mammal.
The above applications of a composition may be embodied in a method comprising administering a composition comprising red microalgae cell material (such as a homogenate) for ingestion by a mammal as described herein. In some cases, the composition comprises one or more other agents or compounds with anti-oxidant activity.
VII Gene Expression in Microalgae
Genes can be expressed in microalgae by providing, for example, coding sequences in operable linkage with promoters.
An exemplary vector design for expression of a gene in microalgae contains a first gene in operable linkage with a promoter active in algae, the first gene encoding a protein that imparts resistance to an antibiotic or herbicide. Optionally the first gene is followed by a 3' untranslated sequence containing a polyadenylation signal. The vector may also contain a second promoter active in algae in operable linkage with a second gene. The second gene can encode any protein, for example an enzyme that produces small molecules or a mammalian growth hormone that can be advantageously present in a nutraceutical.
It is preferable to use codon-optimized cDNAs: for methods of recoding genes for expression in microalgae, see for example US patent application 20040209256.
It has been shown that many promoters in expression vectors are active in algae, including both promoters that are endogenous to the algae being transformed algae as well as promoters that are not endogenous to the algae being transformed (ie: promoters from other algae, promoters from plants, and promoters from plant viruses or algae viruses). Example of methods for transforming microalgae, in addition to those demonstrated in the Examples section below, including methods comprising the use of exogenous and/or endogenous promoters that are active in microalgae, and antibiotic resistance genes functional in microalgae, have been described. See for example; Curr Microbiol. 1997 Dec;35(6):356-62 (Chlorella vulgaris); Mar Biotechnol (NY). 2002 Jan;4(l):63-73 (Chlorella ellipsoidea); MoI Gen Genet. 1996 Oct 16;252(5):572-9 (Phaeodactylum tricornutum); Plant MoI Biol. 1996 Apr;31(1): 1-12 (Volvox carteri); Proc Natl Acad Sci U S A. 1994 Nov 22;91(24):11562-6 (Volvox carteri); Falciatore A, Casotti R, Leblanc C, Abrescia C, Bowler C, PMID: 10383998, 1999 May;l(3):239-251 (Laboratory of Molecular Plant Biology, Stazione Zoologjca, Villa Comunale, 1-80121 Naples, Italy) (Phaeodactylum tricornutum and Thalassiosira weissflogii); Plant Physiol. 2002 May;129(l):7-12. (Porphyridium sp.); Proc Natl Acad Sci U S A. 2003 Jan 21;100(2):438-42. (Chlamydomonas reinhardtii); Proc Natl Acad Sci U S A. 1990 Feb;87(3): 1228-32. (Chlamydomonas reinhardtii); Nucleic Acids Res. 1992 Jun 25;20(12):2959-65; Mar Biotechnol (NY). 2002 Jan;4(l):63-73 (Chlorella); Biochem MoI Biol Int. 1995 Aug;36(5):1025-35 (Chlamydomonas reinhardtii); J Microbiol. 2005 Aug;43(4):361-5 (Dunaliella); Yi Chuan Xue Bao. 2005 Apr;32(4):424-33 (Dunaliella); Mar Biotechnol (NY). 1999 May;l(3):239-251. (Thalassiosira and Phaedactylum); Koksharova, Appl Microbiol Biotechnol 2002 Feb;58(2): 123-37 (various species); MoI Genet Genomics. 2004 Feb;271(l):50-9 (Thermosynechococcus elongates); J. Bacterid. (2000), 182, 211-215; FEMS Microbiol Lett. 2003 Apr 25;221(2): 155-9; Plant Physiol. 1994 Jun; 105(2):635-41; Plant MoI Biol. 1995 Dec;29(5):897-907 (Synechococcus PCC 7942); Mar Pollut Bull. 2002;45(l-12):163~7 (Anabaena PCC 7120); Proc Natl Acad Sci U S A. 1984 Mar;81(5):1561-5 (Anabaena (various strains)); Proc Natl Acad Sci U S A. 2001 Mar 27;98(7):4243-8 (Synechocystis); Wirth, MoI Gen Genet 1989 Mar;216(l):175-7 (various species); MoI Microbiol, 2002 Jun;44(6): 1517-31 and Plasmid, 1993 Seρ;30(2):90-105 (Fremyella diplosiphon); Hall et al. (1993) Gene 124: 75-81 (Chlamydomonas reinhardtii); Gruber et al. (1991). Current Micro. 22: 15-20; Jarvis et al. (1991) Current Genet. 19: 317- 322 (Chlorella); for additional promoters see also Table 1 from US Patent 6,027,900). For additional methods of transforming microalgae of the genus Porphyridium, see PCT application number WO2006013572, entitled "RED MICROALGAE EXPRESSING EXOGENOUS POLYPEPTIDES AND METHODS OF GENERATING AND UTILIZING SAME", Application number WO2005IL00842 20050804, priority number(s) US20040598849P 20040805, which includes use of a cauliflower mosaic virus (CMV) promoter (See SEQ ID NO:45). Also see SEQ ID NO:44, a promoter that drives the gene encoding the glycoprotein that binds the polysaccharide from Porphyridium sp. (see GenBank accession number AY778963 for the cDNA of this gene). The promoter of SEQ ID NO:44 is a preferred promoter for driving exogenous genes in algae of the genus Porphyridium.
Suitable promoters may be used to express a nucleic acid sequence in microalgae. In some embodiments, the sequence is that of an exogenous gene or nucleic acid. In particular embodiments, the exogenous gene is one that encodes a carbohydrate transporter protein. Such a gene may be advantageously expressed in a microalgal cell to allow entry of a monosaccharide transported by the transporter protein. Tn other embodiments, the exogenous gene can encode a superoxide dismutase or a mammalian growth hormone. In cases of an exogenous nucleic acid coding sequence, the codon usage may be optionally optimized in whole or in part to facilitate expression in microalgae. In some embodiments the invention includes cells of the genus Porphyridium that have been stably transformed with a vector containing a selectable marker gene in operable linkage with a promoter active in microalgae. In other embodiments the invention includes cells of the genus Porphyridium that have been stably transformed with a vector containing a- selectable marker gene in operable linkage with a promoter endogenous to a member of the Rhodophyte phylum. Such promoters include SEQ ID NOs: 44 and 55, promoters from the genome of Chondrus crispus (Genbank accession number Z47547), promoters from the genome of Cyanidioschyzon merolae (see for example Matsuzaki, M. et al. Nature 428, 653-657 (2004); Plant Physiology 137:567-585 (2005); entire sequence available at http://merolae.biol. s.u-tokvo.ac.jp/db/chromosome.cgi). In other embodiments the invention includes cells of the genus Porphyridium that have been stably transformed with a vector containing a selectable marker gene in operable linkage with a promoter other than a CMV promoter such as that found in PCT application WO2006013572. The invention thus includes, in some embodiments, a microalgal cell comprising an exogenous gene that encodes a carbohydrate transporter protein. The cell may be that of the genus Porphyridum as a non-limiting example. Non-limiting examples of genes encoding carbohydrate transporters to facilitate the uptake of exogenously provided carbohydrates include genes encoding the proteins of SEQ ID NOs: 20, 22, 24, 26, 27, 29-39 and 46-48 as provided herein. In some embodiments the nucleic acid sequence encodes a protein with at least 60% amino acid sequence identity with a protein with a sequence represented by one of SEQ ID NOs: 20, 22, 24, 26, 27, and 29-39 and 46-48 wherein the protein is located in the plasma membrane of the cell and transports a carbohydrate from the culture media into the cell. In other embodiments, the nucleic acid sequence encodes a protein with at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 98%, or higher, amino acid identity with a sequence of these SEQ ID NOs: 20, 22, 24, 26 and 27. In further embodiments, the nucleic acid sequence has at least 60% nucleotide identity with a nucleic acid molecule with a one of SEQ ED NOs: 21, 23 and 25. In other embodiments, the nucleic acid sequence has at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 98%, or higher, nucleic acid identity with a sequence of these SEQ ID NOs.
Hexokinases can also be expressed to facilitate utilization of exogenously provided carbohydrates through phosphorylation of monosaccharides. A nonlimiting example of a hexokinase is SEQ IS NO: 71 or a protein with at least 60% amino acid identity with SEQ ID NO:71 that phosphorylates a monosaccharide.
In other embodiments, the invention provides for the expression of a protein sequence found to be tightly associated with microalgal polysaccharides. One non-limiting example is the protein of SEQ ID NO: 28, which has been shown to be tightly associated with, but not covalently bound to, the polysaccharide from Porphyridium sp. (see J. Phycol. 40: 568-580 (2004)). When Porphyridium culture media is subjected to tangential flow filtration using a filter containing a pore size well in excess of the molecular weight of the protein of SEQ ID NO: 28, the polysaccharide in the retentate contains detectable amounts of the protein, indicating its tight association with the polysaccharide. The calculated molecular weight of the protein is approximately 58kD, however with glycosylation the protein is approximately 66 kD.
Such a protein may be expressed directly such that it will be present with the polysaccharides of the invention or expressed as part of a fusion or chimeric protein as described herein. As a fusion protein, the portion that is tightly associated with a microalgal polysaccharide effectively links the other portion(s) to the polysaccharide. A fusion protein may comprise a second protein or polypeptide, with a homogenous or heterologous sequence. A homogenous sequence would result in a dimer or multimer of the protein while a heterologous sequence can introduce a new functionality, including that of a bioactive protein or polypeptide.
Non-limiting examples of the second protein include an antibody, a growth hormone or factor, and an enzyme. In optional embodiments, the enzyme is superoxide dismutase, such as that has at least about 60% amino acid identity with the sequence of SEQ ID NO: 14 or SEQ ID NO: 15 as non-limiting examples. Superoxide dismutase scavenges reactive oxygen species such as the superoxide anion. In some embodiments, the superoxide dismutase has at least about 70%, at least about 75%, at least about 80%,. at least about 85%, at least about 90%, at least about 95%, or at least about 98%, or higher, amino acid identity with the sequence of SEQ ID NO: 14 or 15 or a sequence from Table 16. In other embodiments, the enzyme is a phytase (such as GenBank accession number CAB91845 and US Patents 6,855,365 and 6,110,719).
A fusion between the polysaccharide binding protein and antibodies that specifically bind to and neutralize a pathogen are included in the invention. Non-limiting examples include anti-HIV antibodies, like the 2Gl 2 antibody (see Proc Natl Acad Sci U S A. 2005 Sep 20;102(38): 13372-7); the 1RHH_B antibody (see Clin Exp Immunol. 2005 Jul;141(l):72-80); the scFvlO2 antibody (see J Gen Virol. 2005 Jun;86(Pt 6):1791-800); and the micro Ab antibody (see Nat Med. 2005 Jun;l l(6):615-22; 2Gl 2, 2F5, 4E10, 2gl2 Fab 1ZLS_L). These and other antibodies, preferably antibodies that specifically bind to infectious disease agents, can also be expressed in algae without being fused to any other proteins. The biomass containing the recombinant antibodies can be administered orally to deliver the antibodies to a mammal for prophylaxis or treatment.
One advantage to a fusion is that the bioactivity of the polysaccharide and the bioactivity from the protein can be combined in a product without increasing the manufacturing cost over only purifying the polysaccharide. As a non-limiting example, the potent antioxidant properties of a Porphyridium polysaccharide can be combined with the potent antioxidant properties of superoxide dismutase in a fusion, however the polysaccharide:superoxide dismutase combination can be isolated to a high level of purity using tangential flow filtration. In another non-limiting example, the potent antiviral properties of s. Porphyridium polysaccharide can be added to the potent neutralizing activity of recombinant antibodies fused to the protein (SEQ ID NO:28) that tightly associates with the polysaccharide. In a preferred embodiment, fusion proteins of SEQ ID NO:28 or sequences with at least 60% amino acid identity with SEQ ID NO:28 bind with high affinity to a sulfated exopoly saccharide from a cell of the genus Porphyridium. It is preferred but not becessary that the binding be selective.
In other embodiments, the exogenous gene can encode a protein that catalyzes the conversion of one carotenoids/xanthophyll to another, such as a beta-carotene hydroxylase (beta carotene to zeaxanthin), a lycopene cyclase (lycopene to beta carotene), and other enzymes known in the art. In cases of an exogenous nucleic acid coding sequence, the codon usage may be optionally optimized in whole or in part to facilitate expression in microalgae. Beta carotene hydroxylases exhibit a wide degree of amino acid identity despite the fact that that are capable of catalyzing the same reaction. The invention includes cloning and expression of enzymes that catalyze the conversion of beta carotene to zeaxanthin that have at least 25% amino acid identity to those in Table 4. Optionally, the beta carotene hydroxylases of the invention have at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, and at least 99% amino acid identity to those in Table 4.
The invention includes cloning and expression of enzymes that catalyze the conversion of lycopene to beta carotene (ie: lycopene cylases). Optionally, the lycopene cylases of the invention have at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, and at least 99% amino acid identity to GenBank accession numbers AAA81880, NP_624526, BAA20275, and P21687.
Beta carotene hydroxylases exhibit widely divergent amino acid identity while exhibiting the same catalytic activity. For example, BLAST comparisons of beta carotene hydroxylases show as little as 25% amino acid identity: NP_682690 vs. NP_896386 (59% identity); ABB49299 vs. YP_172377 (67% identity); NP_682690 vs. AAT38625 (25% identity); YP_457405 vs. NP_440788 (25% identity); ZPJ)1084736 vs. NP_848964 (55% identity). BLAST parameters used in the analysis were default parameters (blastp program, matrix BLOSUM62, open gap 11 and extension gap I9 gapx_dropoff 50, expect 10.0, word size 3, filter activated).
In other embodiments, the invention includes genetic expression methods comprising the use of an expression vector. In one method, a microalgal cell, such as a Porphyridium cell, is transformed with a dual expression vector under conditions wherein vector mediated gene expression occurs. The expression vector may comprise a resistance cassette comprising a gene encoding a protein that confers resistance to an antibiotic, such as zeocin, or another selectable marker such as a carbohydrate transporter gene for selection in the dark in the presence of a fixed carbon source, operably linked to a promoter active in microalgae. The vector may also comprise a second expression cassette comprising a second protein to a promoter active in microalgae. The two cassettes are physically linked in the vector. The transformed cells may be optionally selected based upon the ability to grow in the presence of the antibiotic or other selectable marker under conditions wherein cells lacking the resistance cassette would not grow, such as in the dark. The resistance cassette, as well as the expression cassette, may be taken in whole or in part from another vector molecule.
Tn one non-limiting example, a method of expressing an exogenous gene in a cell of the genus Porphyridium is provided. The method may comprise operably linking a gene encoding a protein that confers resistance to the antibiotic zeocin to a promoter active in microalgae to form a resistance cassette; operably linking a gene encoding a second protein to a promoter active in microalgae to form a second expression cassette, wherein the resistance cassette and second expression cassette are physically connected to form a dual expression vector; transforming the cell with the dual expression vector; and selecting for the ability to survive in the presence of at least 2.5 ug/ml zeocin, preferably at least 3.0 ug/ml zeocin., and more preferably at least 3.5 ug/ml zeocin, more preferably at least 5.0 ug/ml zeocin.
In some embodiments, the expression cassette expresses a growth hormone, optionally mammalian and optionally a fish or other vertebrate growth hormone. Non- limiting examples of the growth hormone include bovine growth hormone, human growth hormone, porcine growth hormone, and equine growth hormone. Other non-limiting examples growth hormones include proteins with sequences represented as SEQ ID NOs: 17, 18 and 19. In other embodiments, the growth hormone has at least about 60%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 98%, or higher, amino acid identity with a protein sequence represented by one of SEQ ID NOs: 17, 18, 19 or one of Genbank accession numbers AAW67479; AAW67480; AAB641 17; BAC07248; AAF61751; AAX77220; ABA55647; ABI97975; BAA06379; AAU93606; AAX31661; NP_001003168, further wherein the protein stimulates growth in a vertebrate organism. In another embodiment the expression cassette expresses a phytase enzyme, such as for example, SEQ ID NOs: 40 and 41.
In other aspects, the invention provides the cells, such as Porphyridium cells, prepared by the above methods. The cells, whether in whole or extracted or homogenized form, may comprise a mammalian growth hormone via recombinant protein expression as provided above. In a preferred embodiment, transgenic Porphyridiwn expressing a mammalian growth hormone and /or a phytase enzyme are formulated into livestock food and administered to animals.
The cells may be advantageously used as, or as a component of, animal feed. The advantage to expressing such growth hormones in microalgae such as Porphyridium is that the polysaccharide is not hydro lyzed by the stomach of the mammal (see for example Br J Nutr. 2000 Oct;84(4):469-76), and therefore the cell wall, which is made primarily of polysaccharide, protects the mammalian growth hormone as the cells transit through the stomach and into the intestines. Once in the intestines, the cell wall eventually begins breaking down, allowing the growth hormones to cross into the bloodstream and achieve a pharmacological effect.
In additional aspects, the expression of a protein that produces small molecules in microalgae is included and described. Some genes that can be expressed using the methods provided herein encode enzymes that produce nutraceutical small molecules such as lutein, zeaxanthin, and DHA. Preferably the genes encoding the proteins are synthetic and are created using preferred codons on the microalgae in which the gene is to be expressed. For example, enzyme capable of turning EPA into DHA are cloned into the microalgae Porphyridium sp. by recoding genes to adapt to Porphyridium sp. preferred codons. For examples of such enzymes see Nat Biotechnol. 2005 Aug;23(8):1013-7. For examples of enzymes in the carotenoid pathway see SEQ ID NOs: 12 and 13 and sequences from Table 15. The advantage to expressing such genes is that the nutraceutical value of the cells increases without increasing the manufacturing cost of producing the cells.
For sequence comparison to determine percent nucleotide or amino acid identity, typically one sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are input into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. The sequence comparison algorithm then calculates the percent sequence identity for the test sequence(s) relative to the reference sequence, based on the designated program parameters. Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith & Waterman, Adv. Appl. Math. 2:482 (1981), by the homology alignment algorithm of Needleman & Wunsch, J. MoI. Biol. 48:443 (1970), by the search for similarity method of Pearson & Lipman, Proc. Nat 'I. Acad. Sci. USA 85:2444 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, WI), or by visual inspection (see generally Ausubel et ah, supra). ■
Another example of algorithm that is suitable for determining percent sequence identity and sequence similarity is the BLAST algorithm, which is described in Altschul et ah, J. MoI. Biol. 215:403-410 (1990). Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (http://www.ncbi.nlm.nih.gov/). This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul et ah, supra.). These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always > 0) and N (penalty score for mismatching residues; always < 0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. For identifying whether a nucleic acid or polypeptide is within the scope of the invention, the default parameters of the BLAST programs are suitable. The BLASTN program (for nucleotide sequences) uses as defaults a word length (W) of 11, an expectation (E) of 10, M=5, N=-4, and a comparison of both strands. For amino acid sequences, the BLASTP program uses as defaults a word length (W) of 3, an expectation (E) of 10, and the BLOSUM62 scoring matrix. The TBLATN program (using protein sequence for nucleotide sequence) uses as defaults a word length (W) of 3., an expectation (E) of 10, and a BLOSUM 62 scoring matrix, (see Henikoff & Henikoff, Proc. Natl. Acad. ScL USA 89:10915 (1989)).
In addition to calculating percent sequence identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin & Altschul, Proc. Nat'l. Acad. ScL USA 90:5873-5787 (1993)). One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001.
VIII Methods of Trophic Conversion
As explained herein, microalgae generally have the ability to live off a fixed carbon sources such as glucose, but many do not have transporters that allow for uptake of the fixed carbon source from the culture media. Microalgae cells can be transformed with a gene that encodes a plasma membrane sugar transporter that allows for the selection of growth in the dark, in the absence of photosynthesis, in the presence of the transporter's substrate sugar. Such transformed cells provide a significant benefit in that the need for light energy is reduced or eliminated because the cells may grow and produce cellular products, including polysaccharides, in the presence of fixed carbon material as the energy source. See for example, Science. 2001 Jun 15;292(5524):2073-5. Such growth achieves much higher cell densities in a shorter period of time than photoautotrophic growth.
The transformed microalgal cell may be one that is described above as expressing a sugar transporter. Nucleic acids and vectors for such expression are also described above. For example, nucleic acids encoding carbohydrate transporters such as SEQ ID NOs: 20, 22, 24, 26, 27, 29-39 and 46-48 are placed in operable linkage with a promoter active in microalgae. Preferably, the nucleic acid encoding a carbohydrate transporter contains preferred codons of the organism the vector is transformed into. For example, the nucleic acids of SEQ ID NOs: 21, 23, and 25 encode the carbohydrate transporter proteins of SEQ ID NOs: 20, 22, and 24, respectively. As a nonlimiting example, a codon-optimized cDNA encoding a carbohydrate transporter protein, optimized for expression in Porphyridium sp., is placed in operable linkage with a promoter and 3'UTR active in microalgae. The vector is used to transform a cell of the genus Porphyridium using methods disclosed herein, including biolistic transformation, electroporation, and glass bead transformation. A preferred promoter is active in more than one species of microalgae, such as for example the Chlamydomonas reinhardtii RBCS2 promoter (SEQ ID NO:43). Any promoter active in microalgae can be used to express a gene in such constructs, and preferred promoters such as RBCS2 and viral promoters have been shown to be active in multiple species of microalgae (see for example Plant Cell Rep. 2005 Mar;23(10-l l):727-35; J Microbiol. 2005 Aug;43(4):3όl-5; Mar Biotechnol (NY). 2002 Jan;4(l):63-73). Promoters, cDNAs, and 3'UTRs, as well as other elements of the vectors, can be generated through cloning techniques using fragments isolated from native sources (see for example Molecular Cloning: A Laboratory Manual, Sambrook et al. (3d edition, 2001, Cold Spring Harbor Press; and U.S. Patent 4,683,202). Alternatively, elements can be generated synthetically using known methods (see for example Gene. 1995 Oct 16;164(l):49-53),
Alternatively, cells may be mutagenized and then selected for the ability to grow in the absence of light energy but in the presence of a fixed carbon source.
Thus the invention includes a method of producing microalgal cells that have gained the ability to grow via a fixed carbon source in the absence of photosynthesis. This may also be referred to as trophic conversion of a microalgal cell to no longer be an obligate photoautotroph. In some embodiments, the method comprises identifying or selecting cells that have gained the ability to utilize energy from a fixed carbon source.
In some embodiments, the methods comprise selecting microalgal cells, such as a Porphyridium cell, for the ability to undergo cell division in the absence of light, or light energy. The cells, such as one from a species listed in Table 1 , may be those which have been transformed with a sugar transporter or those which have been mutagenized, chemically or non-chemically. The selection may be, for example, on about 0.1% or about 1% glucose, or another fixed carbon source, in the dark. Preferred fixed carbon compounds are listed in Tables 2 and 3.
Non-limiting examples of carbohydrate transporter proteins, optionally operably linked to promoters active in microalgae, as well as expression cassettes and vectors comprising them, have been described above. Alternatively, the nucleic acids may be incorporated into the genome of a microalgal cell such that an endogenous promoter is used to express the transporter. Additional embodiments of the methods include expression of transporters of a carbohydrate selected from Table 2 or 3. Non-limiting examples of mutagenesis include contact or propagation in the presence of a mutagen, such as ultraviolet light, nitrosoguanidine, and/or ethane methyl sulfonate (EMS). As one non-limiting example, a method of the invention comprises providing a nucleic acid encoding a carbohydrate transporter protein; transforming a Porphyridium cell with the nucleic acid; and selecting for the ability to undergo cell division in the absence of light or in the presence of a carbohydrate that is transported by the carbohydrate transporter protein. In another non-limiting example, a method comprises subjecting a microalgal cell to a mutagen; placing the cell in the presence of a molecule listed in Tables 2 or 3; and selecting for the ability to undergo cell division in the absence of light.
The methods may also be considered to be for trophically converting a microalgal cell to no longer be an obligate phototroph. It is pointed out that the ability to select for loss of obligate phototrophism also provides an alternative means to select for expression of a sugar transporter in the absence of a selectable marker because correct expression and functionality of the transporter is the selectable phenotype when cells are grown in the absence of light for photosynthesis.
It should be apparent to one skilled in the art that various embodiments and modifications may be made to the invention disclosed in this application without departing from the scope and spirit of the invention. All publications mentioned herein are cited for the purpose of describing and disclosing reagents, methodologies and concepts that may be used in connection with the present invention. Nothing herein is to be construed as an admission that these references are prior art in relation to the inventions described herein.
EXAMPLE 1
Growth of Porphyridium cruentum and Porphyridium sp.
Porphyridium sp. (strain UTEX 637) and Porphyridium cruentum (strain UTEX 161) were inoculated into autoclaved 2 liter Erlenmeyer flasks containing an artificial seawater media: 1495 ASW medium recipe from the American Type Culture Collection, (components are per 1 liter of media) NaCl: 27.Og; MgSO4 • 7H2O: 6.6g; MgCl2 . 6H2O: 5.6g; CaCl2 . 2H2O: 1.5g; KNO3: 1.Og; KH2PO4 : 0.07g; NaHCO3: 0.04 g; 1.0 M Tris-HCl buffer, pH 7.6:20.0 ml; Trace Metal Solution (see below): 1.0 ml; Chelated Iron Solution (see below): 1.0 ml; Distilled water: bring-to 1.0 L. Trace Metal Solution: ZnCl2:4.0 mg; H3BO3: 60.0 mg; CoCl2 . 6H2O: 1.5 mg; CuC12 . 2H2O: 4.0 mg; MnCl2 . 4H2O: 40.0 mg; (NH4^Mo7O24 . 4H2O: 37.0 mg; Distilled water: 100.0 ml. Chelated Iron Solution: FeCl3 . 4H2O: 240.0 mg; 0.05 M EDTA, pH 7.6: 100.0 ml. Media was autoclaved for at least 15 minutes at 1210C. Inoculated cultures in 2 liter flasks were maintained at room temperature on stir plates. Stir bars were placed in the flasks before autoclaving. A mixture of 5% CO2 and air was bubbled into the flasks. Gas was filter sterilized before entry. The flasks were under 24 hour illumination from above by standard fluorescent lights (approximately 150 uE/m"1 /s"1). Cells were grown for approximately 12 days, at which point the cultures contained approximately of 4 X 106 cells/mL.
EXAMPLE 2
Dense Porphyridium sp. and Porphyridium omentum cultures were centrifuged at 4000 rcf. The supernatant was subjected to tangential flow filtration in a Millipore Pellicon 2 device through a 100OkD regenerated cellulose membrane (filter catalog number P2C01MC01). Approximately 4.1 liters of Porphyridium cruentum and 15 liters of Porphyridium sp. supernatants were concentrated to a volume of approximately 200 ml in separate experiments. The concentrated exopolysaccharide solutions were then diafiltered with 10 liters of ImM Tris (pH 7.5). The retentate was then flushed with ImM Tris (pH 7.5), and the total recovered polysaccharide was lyophilized to completion. Yield calculations were performed by the dimethylmethylene blue (DMMB) assay. The lyophilized polysaccharide was resuspended in deionized water and protein was measured by the bicinchoninic acid (BCA) method. Total dry product measured after lyophilization was 3.28g for Porphyridium sp. and 2.Og for Porphyridium cruentum. Total protein calculated as a percentage of total dry product was 12.6% for Porphyridium sp. and 15.0% for Porphyridium cruentum.
EXAMPLE 3
A measured mass (approximately 125 grams) of freshly harvested Porphyridium sp. cells, resuspended in a minimum amount of dHaO sufficient to allow the cells to flow as a liquid, was placed in a container. The cells were subjected to increasing amounts of sonication over time at a predetermined sonication level. Samples were drawn at predetermined time intervals, suspended in measured volume of dH2O and diluted appropriately to allow visual observation under a microscope and measurement of polysaccharide concentration of the cell suspension using the DMMB assay. A plot was made of the total amount of time for which the biomass was sonicated and the polysaccharide concentration of the biomass suspension. Two experiments were conducted with different time intervals and total time the sample was subjected to sonication. The first data set from sonication experiment 1 was obtained by subjecting the sample to sonication for a total time period of 60 minutes in 5 minute increments. The second data set from sonication experiment 2 was obtained by subjecting the sample to sonication for a total time period of 6 minutes in 1 -minute increments. The data, observations and experimental details are described below. Standard curves were generated using TFF-purifϊed, lyophilized, weighed, resuspended Porphyridium sp. exopolysaccharide.
General parameters of sonication experiments 1 and 2
Cells were collected and volume of the culture was measured. The biomass was separated from the culture solution by centrifugation. The centrifuge used was a Forma Scientific Centra-GP8R refrigerated centrifuge. The parameters used for centrifugation were 4200rpm, 8 minutes, rotor# 218. Following centrifugation, the biomass was washed with dH2θ. The supernatant from the washings was discarded and the pelleted cell biomass was collected for the experiment.
A sample of lOOμL of the biomass suspension was collected at time point 0 (OTP) and suspended in 900μL dH2O. The suspension was further diluted ten-fold and used for visual observation and DMMB assay. The time point 0 sample represents the solvent- available polysaccharide concentration in the cell suspension before the cells were subjected to sonication. This was the baseline polysaccharide value for the experiments.
The following sonication parameters were set: power level=8, 20 seconds ON / 20 seconds OFF (Misonix 3000 Sonicator with flat probe tip). The container with the biomass was placed in an ice bath to prevent overheating and the ice was replenished as necessary. The sample was prepared as follows for visual observation and DMMB assay: 100 μL of the biomass sample + 900μL dH2Q was labeled as dilution 1. 100 μL of (i) dilution 1 + 900μL dH^O for cell observation and DMMB assay.
Sonication Experiment 1
In the first experiment the sample was sonicated for a total time period of 60 minutes, in 5-minute increments (20 seconds ON / 20 seconds OFF). The data is presented in Tables 4, 5 and 6. The plots of the absorbance results are presented in Figure 5.
TABLE 4: SONICATION RECORD - EXPERIMENT 1
TP = time point.
TABLE 5: STANDARD CURVE RECORD - - SONICATION EXPERIMENT 1
Table 6: Record of Sample Absorbance versus Time Points - Sonication Experiment 1
The plot of polysaccharide concentration versus sonication time points is displayed above and in Figure 5, Solvent-available polysaccharide concentration of the biomass (cell) suspension reaches a maximum value after 5 minutes of sonication. Additional sonication in 5-minute increments did not result in increased solvent-available polysaccharide concentration.
Homogenization by sonication of the biomass resulted in an approximately 10-fold increase in solvent-available polysaccharide concentration of the biomass suspension, indicating that homogenization significantly enhances the amount of polysaccharide available to the solvent. These results demonstrate that physically disrupted compositions of P 'orphyridium for oral or other administration provide novel and unexpected levels or polysaccharide bioavailability compared to compositions of intact cells. Visual observation of the samples also indicates rupture of the cell wall and thus release of insoluble cell wall- bound polysaccharides from the cells into the solution that is measured as the increased polysaccharide concentration in the biomass suspension. Sonication Experiment 2
In the second experiment the sample was sonicated for a total time period of 6 minutes in 1 -minute increments. The data is presented in Tables 7, 8 and 9. The plots of the absorbance results are presented in Figure 6.
TABLE 7: SONICATION EXPERIMENT 2
TABLE 8: STANDARD CURVERECORD- SONICATION EXPERIMENT2
Table 9: Record of Sample Absorbance versus Time Points — Sonication Experiment 2
The value of the solvent-available polysaccharide increases gradually up to the 5 minute time point as shown in Table 9 and Figure 6.
EXAMPLE 4 Porphyridium sp. culture was centrifuged at 4000 rcf and supernatant was collected. The supernatant was divided into six 30ml aliquots. Three aliquots were autoclaved for 15 min at 1210C. After cooling to room temperature, one aliquot was mixed with methanol (58.3% vol/vol), one was mixed with ethanol (47.5% vol/vol) and one was mixed with isopropanol (50% vol/vol). The same concentrations of these alcohols were added to the three supernatant aliquots that were not autoclaved. Polysaccharide precipitates from all six samples were collected immediately by centrifugation at 4000 rcf at 2O0C for 10 min and pellets were washed in 20% of their respective alcohols. Pellets were then dried by lyophilization and resuspended in 15 ml deionized water by placement in a 600C water bath. Polysaccharide pellets from non-autoclaved samples were partially soluble or insoluble. Polysaccharide pellets from autoclaved ethanol and methanol precipitation were partially soluble. The polysaccharide pellet obtained from isopropanol precipitation of the autoclaved supernatant was completely soluble in water.
EXAMPLE S
Approximately 10 milligrams of purified polysaccharide from Porphyridium sp. and Porphyridium cruentum (described in Example 3) were subjected to monosaccharide analysis.
Monosaccharide analysis was performed by combined gas chromatography/mass spectrometry (GC/MS) of the per-O-trimethylsilyl (TMS) derivatives of the monosaccharide methyl glycosides produced from the sample by acidic methanolysis.
Methyl glycosides prepared from 500μg of the dry sample provided by the client by methanolysis in 1 M HCl in methanol at 80°C (18-22 hours), followed by re-N-acetylation with pyridine and acetic anhydride in methanol (for detection of amino sugars). The samples were then per-O-trimethylsilylated by treatment with Tri-Sil (Pierce) at 800C (30 mins). These procedures were carried out as previously described described in Merkle and Poppe (1994) Methods Enzymol. 230: 1-15; York, et al. (1985) Methods Enzymol. 118:3- 40. GC/MS analysis of the TMS methyl glycosides was performed on an HP 5890 GC interfaced to a 5970 MSD, using a Supelco DB-I fused silica capillary column (30m 0.25 mm ID).
Table 10: Porphyridium sp. monosaccharide analysis
Glycosyl residue Mass (μg) Mole %
Arabinose (Ara) n.d. n.d. Rhamnose (Rha) 2.7 1.6
Fucose (Fuc) n.d. n.d.
Xylose (XyI) 70.2 44.2
Glucuronic acid (GIcA) n.d. n.d.
Galacturonic acid (GaIA) n.d. n.d.
Mannose (Man) 3.5 1.8
Galactose (Gal) 65.4 34.2
Glucose (GIc) 34.7 18.2
JV-acetyl galactosamine (GaINAc) n.d. n.d.
7V-acetyl glucosamine (GIcNAc) trace trace
∑=176.5
Table 11 : Porphyridium cruentum monosaccharide analysis Glycosyl residue Mass (μg) Mole %
Arabinose (Ara) n.d. ■ n.d.
Rhamnose (Rha) n.d. n.d.
Fucose (Fuc) n.d. n.d.
Xylose (XyI) 148.8 53.2
Glucuronic Acid (GIcA) 14.8 4.1
Mannose (Man) n.d. n.d.
Galactose (Gal) 88.3 26.3
Glucose (GIc) 55.0 16.4
N-acetyl glucosamine (GIcNAc) trace trace
N-acetyl neuraminic acid (NANA) n.d. n.d.
Σ= 292.1
Mole % values are expressed as mole percent of total carbohydrate in the sample. n.d.= none detected.
EXAMPLE 6
Porphyridium sp. was grown as described. 2 liters of centrifuged Porphyridium sp. culture supernatant were autoclaved at 121°C for 20 minutes and then treated with 50% • isopropanol to precipitate polysaccharides. Prior to autoclaving the 2 liters of supernatant contained 90.38 mg polysaccharide. The pellet was washed with 20% isopropanol and dried by lyophilization. The dried material was resuspended in deionized water. The resuspended polysaccharide solution was dialyzed to completion against deionized water in a Spectra/Por cellulose ester dialysis membrane (25,000 MWCO). 4.24% of the solid content in the solution was proteins as measured by the BCA assay.
EXAMPLE 7
Porphyridium sp. was grown as described. 1 liters of centrifuged Porphyridium sp. culture supernatant was autoclaved at 1210C for 15 minutes and then treated with 10% protease (Sigma catalog number P-5147; protease treatment amount relative to protein content of the supernatant as determined by BCA assay). The protease reaction proceeded for 4 days at 37°C. The solution was then subjected to tangential flow filtration in a Millipore Pellicon® cassette system using a 0.1 micrometer regenerated cellulose membrane. The retentate was diafiltered to completion with deionized water. No protein was detected in the diafiltered retentate by the BCA assay. See Figure 8.
Optionally, the retentate can be autoclaved to achieve sterility if the filtration system is not sterile. Optionally the sterile retentate can be mixed with pharmaceutically acceptable carrier(s) and filled in vials for injection.
Optionally, the protein free polysaccharide can be fragmented by, for example, sonication to reduce viscosity for parenteral injection as, for example, an antiviral compound. Preferably the sterile polysaccharide is not fragmented when prepared for injection as a joint lubricant.
EXAMPLE 8
Cultures of Porphyridium sp. (UTEX 637) and Porphyridium cruentum (strain UTEX 161) were grown, to a density of 4 X 106cells/mL, as described in Example 1. For each strain, about 2 X 106 cells/mL cells per well (-500 uL) were transferred to 11 wells of a 24 well microliter plate. These wells contained ATCC 1495 media supplemented with varying concentration of glycerol as follows: 0%, 0.1%, 0.25%, 0.5%, 0.75%, 1%, 2 %, 3%, 5%, 7% and 10%. Duplicate microtiter plates were shaken (a) under continuous illumination of approximately 2400 lux as measured by a VWR Traceable light meter (cat # 21800-014), and (b) in the absence of light. After 5 days, the effect of increasing concentrations of glycerol on the growth rate of these two species of Porphyridium in the light was monitored using a hemocytometer. The results are given in Figure 3 and indicate that in light, 0.25 to 0.75 percent glycerol supports the highest growth rate, with an apparent optimum concentration of 0.5%. Cells in the dark were observed after about 2 weeks of growth. The results are given in Figure 4 and indicate that in complete darkness, 5.0 to 7.0% glycerol supports the highest growth rate, with an apparent optimum concentration of 7.0%.
EXAMPLE 9 Cosmetic Compositions
Porphyridium sp. (UTEX 637) was grown to a density of approximately 4 X 106 cells/mL, as described in Example 1. Approximately 50 grams of wet pelleted, and washed cells were completely homogenized using approximately 20 minutes of sonication as described. The homogenized biomass was mixed with carriers including, water, butylene glycol, mineral oil, petrolatum, glycerin, cetyl alcohol, propylene glycol dicaprylate/dicaprate, PEG-40 stearate, Cl 1-13 isoparaffin, glyceryl stearate, tri (PPG-3 myristyl ether) citrate, emulsifying wax, dimethicone, DMDM hydantoin, methylparaben, carbomer 940, ethylparaben, propylparaben, titanium dioxide, disodium EDTA, sodium hydroxide, butylparaben, and xanthan gum. The mixture was then further homogenized to form a composition suitable for topical administration. The composition was applied to human skin daily for a period of one week.
EXAMPLE 10
Sexually Transmitted Disease Prevention Compositions
Polysaccharide from Porphyridium sp. ws prepared as described in Example 2. Lyophilized polysaccharide was resuspended with distilled water to an antivirally effective concentration of 0.5 milligram per mL. 1.0 mL of the 0.5 mg/mL polysaccharide solution was applied to a latex condom.
In a second composition formulation, 10 microliters of a 1 mg/mL Porphyridium sp. polysaccharide solution was applied to a latex condom. 29 additional 10 microliter increments of the 1 mg/mL solution were successively applied, creating individual sexually transmitted disease composition with between 100 micrograms and 3 milligrams of polysaccharide in 100 microgram increments. See Figure 7.
Other condom formulation techniques can be used (see for example US Patent 6,196,227).
EXAMPLE 11 Approximately 4500 cells (300ul of 1,5x105 cells per ml) of P 'orphyridium sp. and Porphyridium omentum cultures in liquid ATCC 1495 ASW media were plated onto ATCC 1495 ASW agar plates (1.5% agar). The plates contained varying amounts of zeocin, sulfometuron, hygromycin and spectinomycin. The plates were put under constant artificial fluorescent light of approximately 480 lux. After 14 days, plates were checked for growth. Results were as follows; (1) Zeocin: (Concentration μg/ml) [Growth]: (0.0) [+ + + +]; (2.5) [+]; (5.0) [-]; (7.0)[-]. (2) Hygromycin (Concentration μg/ml) [Growth]: (0.0) [ + + + +]; (5.0) [ + + + +]; (10.0) [+ + + +]; (50.0) [+ + + +]. (3) Specinomycin (Concentration μg/ml) [Growth]: (0.0) [+ + + +]; (100.0) [+ + + +]; (250.0) [+ + + +]; (750.0) [+ + + +].
After the initial results above were obtained, a titration of zeocin was performed to more accurately determine growth levels of Porphyridium in the presence of zeocin. Porphyridium sp. cells were plated as described above. Results are shown in Figure 2.
EXAMPLE 12
Trophic Conversion: transporters
Cloning
PIasmid pBluescript KS+ is used as a recipient vector for an expression cassette shown in SEQ ID NO:68. The Porphyridium glycoprotein promoter is cloned into pBluescript KS+, followed by a cauliflower mosaic virus 3' UTR. Unique restriction sites are left between the promoter and 3'UTR. A nucleic acid encoding the glucose transporter of SEQ ID NO:20 using preferred codons of Porphyridium sp. is cloned into the unique restriction sites between the promoter and 3'UTR.
The plasmid is used to transform Porphyridium sp. cells using the biolistic transformation parameters described in Plant Physiol. 2002 May;129(l):7-12. After transformation, some plated cells are scraped from the plate using a sterile cell scraper are transferred into Erlenmeyer flasks wrapped with aluminum foil sufficient to prevent the entry of light into the culture. Identical preparations of transformed, scraped cells are cultured, shaking at -50 rpm in 24 well plates in the dark, in ATCC 1495 media in the presence of 0.1, 1.0, and 2.5% glucose, and monitored for growth. Other cells are transformed on plates containing solid agar ATCC 1495 media, supplemented with either 0.1. 1.0, or 2.5% glucose, and monitored for growth in complete darkness.
EXAMPLE 13 Cultures of P 'orphyridium sp. (UTEX 637) and Porphyridium cruentum (strain UTEX 161) were subjected to chemical mutagenesis (from the protocol in Gorman DS, Levine RP. (1965) Proc Natl Acad Sci U S A. 54(6): 1665-9.). Cells were grown to a density of 4 X 106 cells/mL as described in Example 1. Cells were harvested, washed with 70 mM potassium phosphate buffer (pH 6.9) and resuspended to a density of 4 X 107 cells/mL. To 1 mL of cells (from both strains), 0.1 M ethyl methane sulfonate (EMS) was added. A 200 uL aliquot was taken for the zero time point. The tubes were incubated in the dark at room temperature. 200 uL aliquots were removed from the tube at various time points: 15 min, 30 min, 45 min and 60 min. At each time, the aliquot of cells were treated with 800 uL of 5% sodium thiosulfate to inactivate the EMS. Cells from each aliquot were spun down and washed three times with 1 mL of 70 mM potassium phosphate buffer (pH 6.9), followed by a wash with 1 mL of ATCC 1495 media. The cells were resuspended in 200 uL of ATCC 1495 media, and plated at three different concentrations (IX, 10'2 X, 10"4 X) on duplicate plates of ATCC 1495 media, and incubated under continuous light.
After mutagenesis, some plated cells are scraped from the plate using a sterile cell scraper are transferred into Erlenmeyer flasks wrapped with aluminum foil sufficient to prevent the entry of light into the culture. Identical preparations of transformed, scraped cells are cultured, shaking at ~50 rpm in 24 well plates in the dark, in ATCC 1495 media in the presence of 0.1 , 1.0, and 2.5% glucose, and monitored for growth. Other cells are transformed on plates containing solid agar ATCC 1495 media, with either 0.1, 1.0, or 2.5% glucose, and monitored for growth in complete darkness. Cell treated as described can also be cultured in the presence of an exogenous carbon source from Tables 2 or 3.
EXAMPLE 14
Genetic and nutritional manipulation to generate novel polysaccharides
Cells prepared as described in Example 12 containing a monosaccharide transporter and capable of importing glucose, are cultured in ATCC 1495 media in the light in the presence of 1.0% glucose for approximately 12 days. Exopolysaccharide is purified as described in Example 2. Monosaccharide analysis is performed as described in Example 5.
Cells prepared as described in Example 12 containing a monosaccharide transporter and capable of importing xylose, are cultured in ATCC 1495 media in the light in the presence of 1.0% xylose for approximately 12 days. Exopolysaccharide is purified as described in Example 2. Monosaccharide analysis is performed as described in Example 5. Cells prepared as described in Example 12 containing a monosaccharide transporter and capable of importing galactose, are cultured in ATCC 1495 media in the light in the presence of 1.0% galactose for approximately 12 days. Exopolysaccharide is purified as described in Example 2. Monosaccharide analysis is performed as described in Example 5.
Cells prepared as described in Example 12 containing a monosaccharide transporter and capable of importing glucuronic acid, are cultured in ATCC 1495 media in the light in the presence of 1.0% glucuronic acid for approximately 12 days. Exopolysaccharide is purified as described in Example 2. Monosaccharide analysis is performed as described in Example 5.
Cells prepared as described in Example 12 containing a monosaccharide transporter and capable of importing glucose, are cultured in ATCC 1495 media in the dark in the presence of 1.0% glucose for approximately 12 days. Exopolysaccharide is purified as described in Example 2. Monosaccharide analysis is performed as described in Example 5.
Cells prepared as described in Example 12 containing a monosaccharide transporter and capable of importing xylose, are cultured in ATCC 1495 media in the dark in the presence of 1.0% xylose for approximately 12 days. Exopolysaccharide is purified as described in Example 2. Monosaccharide analysis is performed as described in Example 5.
Cells prepared as described in Example 12 containing a monosaccharide transporter and capable of importing galactose, are cultured in ATCC 1495 media in the dark in the presence of 1.0% galactose for approximately 12 days. Exopolysaccharide is purified as described in Example 2. Monosaccharide analysis is performed as described in Example 5.
Cells prepared as described in Example 12 containing a monosaccharide transporter and capable of importing glucuronic acid, are cultured in ATCC 1495 media in the dark in the presence of 1.0% glucuronic acid for approximately 12 days. Exopolysaccharide is purified as described in Example 2. Monosaccharide analysis is performed as described in Example 5.
EXAMPLE 15
128mg of intact lyophilized Porphyridium sp. cells were ground with a mortar/pestle. The sample placed in the mortar pestle was ground for 5 minutes. 9.0 mg of the sample of the ground cells was placed in a micro centrifuge tube and suspended in lOOOμL of dH2O. The sample was vortexed to suspend the cells. A second sample of 9.0mg of intact, lyophilized Porphyridium sp. cells was placed in a micro centrifuge tube and suspended in 1000μL of dH2O. The sample was vortexed to suspend the cells.
The suspensions of both cells were diluted 1:10 and polysaccharide concentration of the diluted samples was measured by DMMB assay. Upon grinding, the suspension of ground cells resulted in an approximately 10-fold increase in the solvent-accesible polysaccharide as measured by DMMB assay over the same quantity of intact cells.
Table 12
Reduction in the particle size of the lyophilized biomass by homogenization in a mortar/pestle results in better suspension and increase in the solvent-accesible polysaccharide concentration of the cell suspension.
EXAMPLE 16
Porphyridium cruentum was grown as described above in ATCC 1495 media. Porphyridium cruentum culture supernatant were autoclaved at 1210C for 20 minutes. 1.333 liters of isopropanol was added to a 4 liter preparation of autoclaved supernatant to a concentration of 25% (vol/vol). Precipitated exopolysaccharide was removed. Additional isopropanol (381 mL, 786 mL, 167 mL, and 1.333 liters) was added stepwise to the preparation to produce (vol/vol) concentrations of isopropanol of 30%, 38.5%, 40%, and 50%, respectively. Precipitated exopolysaccharide was removed after each increment of isopropanol was added. It was observed that very little additional exopolysaccharide was precipitated upon bringing the concentration from 38.5% to 40% and from 40% to 50%. It was also observed that significant amounts of salt were precipitated upon bringing the concentration from 38.5% to 40% and from 40% to 50%.
An additional 4 liters of exopolysaccharide was precipitated with by addition of 38.5% isopropanol. See Figure 1.
EXAMPLE 17 Preparation of DNA templates for Inverse PCR: Porphyridium sp. genomic DNA (40μg) was digested with Sad at 37°C for 16h in a total volume of 200μl. The digested DNA was extracted with once an equal volume of phenol : chloroform isoamyl alcohol (PCI, 25:24:1) followed by one extraction with equal volume of chloroform. The DNA was precipitated with 1/lOth volume of 3M sodium acetate, pH5.5 and 2.5 volumes of 100% ethanol. The DNA was pelleted by centrifugation, air dried and resuspended in 50μl distilled water to a concentration of 250 ng/ ul. Four dilutions of the DNA (1 μg; 100 ng; 10 ng; 1 ng) was self-ligated for 18h at 16°C in a lOOμl reaction volume using T4 DNA ligase (Invitrogen, Carlsbad, CA). The ligated DNA was extracted once with PCI followed by a chloroform extraction. The sample was ethanol precipitated as described above and resuspended in 200 ul distilled water in 4 individual aliquots at a concentration of 2.5 ng/μL; 250 pg/μL; 25 pg/μL; and 2.5 pg/μL. 1 μL of each of the 4 aliquots was used as template in subsequent inverse PCR reactions.
Inverse PCR: PCR was carried out in 25 μl volume using EXL polymerase (Stratagene, La JoIIa, CA) in 0.2ml thin-walled tubes. PCR reactions were carried out in two rounds. The first round of PCR was set up according to the manufacturer's instructions. The reaction contained 4% DMSO5 1 μL stablilizing solution and 0.25 μM of primers 1 and 2 (primer #1, SEQ ID NO:49, primer #2, SEQ ID NO:50). Inverse PCR primers 1 and 2 were designed in opposite orientation to that of normal PCR and were designed to anneal to the Porphyridium sp. glycoprotein cDNA sequence (GenBank Accession number AY778963). PCR cycle conditions were as follows: initial denaturation of 94°C for 2 minutes followed by 35 cycles of 92°C for 10 seconds, 60°C for 30 seconds and 68°C for 15 minutes, followed by a final extension at 68°C for 5 minutes. The first round PCR product was diluted 100 fold, and 1 μL of the dilution was used as a template for a second round PCR. Second round PCR was done using nested primers 3 and 4 (primer #3, SEQ ID NO:51, primer #4, SEQ ID NO:52). PCR cycle conditions were as follows: initial denaturation of 94°C for 2 minutes followed by 35 cycles of 92°C for 10 seconds, 58°C for 30 seconds and 68°C for 6 minutes, followed by a final extension at 68°C for 5 minutes.
Cloning of the Inverse PCR fragments: A 1.8 kB inverse PCR product was amplified from Sad -digested, self-ligated genomic DNA. The PCR product was cloned into the pGEM-T vector and transformed into JM 109 high efficiency competent cells (Promega, Madison, WI) and selected on LB+ Amp plates containing XGaI/ IPTG. Plasmid DNA extracted from white colonies was screened for inserts and confirmed to contain the 1.8 KB PCR product by sequencing with M13 forward and Ml 3 reverse primers (SEQ ID NOs:53- 54, respectively). Full length sequence of SEQ ID NO:44 was obtained by internal sequencing of the cloned 1.8 kB clone. As expected, the 3' end of the clone was identical to the beginning of the coding region of the glycoprotein cDNA (GenBank Accession number AY778963).
The 1.8kB promoter fragment was amplified by PCR and cloned into operable linkage with a zeocin resistance gene and the carbohydrate transporter cDNAs encoding the proteins of SEQ ID NOs:20, 22, 24, and 46-48 to generate 7 unique Porphyridium transformation vectors.
EXAMPLE 18
The promoter of SEQ ID NO:55 was cloned into operable linkage with a zeocin resitance cDNA encoding the protein of SEQ ID NO: 11. The zeocin resistance cDNA was in operable linkage with the CMV 3' UTR of PCT application WO2006013572. The plasmid was linearized by a restriction enzyme that did not cut in the promoter, cDNA, or 3'UTR.
Porphyridium sp. (strain UTEX 637) culture was grown in ATCC 1495 medium on a gyratory shaker under continuous light at 75 μmol photons m"2 sec"1 until it reached a cell density of 2xl06cells/mL, following which they were incubated in the dark for 24 hours. The dark adapted cells were harvested, and washed twice with sterile distilled water, and resuspended in a Tris-phosphate buffer (2OmM Tris-HCl, pH 7.0; 1 mM potassium phosphate) containing 50 mM sucrose to a density of 4xl08cells/mL. About 250 μL cell suspension (1x108cells) was placed in a disposable electroporation cuvette of 4 mm gap. To the cell suspension, 2-5 μg of linearized plasmid DNA and 200 μg of carrier DNA (sheared salmon sperm DNA) were added. The electroporation cuvette was then incubated in a water bath at 16°C for 10 minutes. An electrical pulse (1800 V/cm) was then applied to the cuvette at a capacitance of 25 μF (no shunt resistor was used for the electroporation) using a Gene Pulser II (Bio-Rad Labs, Hercules, CA) electroporation apparatus. The cuvette was then incubated at room temperature for 5 minutes, following which the cell suspension was transferred to 50 mL of ATCC 1495 media, and shaken on a gyratory shaker for 2 days. Following recovery, the cells were harvested at low speed (4000 rpm), resuspended in distilled water and plated out at low density on solid media (ATCC 1495 + 1% agar) supplemented with 30 μg/mL zeocin (Invitrogen, Carlsbad, CA). The plates were incubated under continuous light at 75 μmol photons m"2 sec"1. Transformants appeared as colonies in 2-3 weeks. EXAMPLE 19 Animal model
The effect of disclosed compositions on total serum cholesterol was assessed with an animal model. Golden Syrian male hamsters weighing between 100 and 120 g were used in this experiment. Animals were randomized into 7 groups of 15 hamsters per group, housed in individual cages and subjected to 12 hr:12 hr lightrdark cycling for 2 week before commencement on the experimental protocol. During the pre-experimental and experimental periods, hamsters were provided with free access to water, and fed ad libitum a semi-synthetic diet (control diet), based on the composition of the AIN-76A which contains 0.25% cholesterol (see Table 13 below).
Table 13 Composition of basal semi-purified diet
Ingredients % (wet weight)
Casein 20.0
Corn starch 28.0
Sucrose 36.3
Lard/safflower oil 5.0
Cellulose 5.0
D-methionine 0.5
Mineral mixture 4.0
Vitamin mixture 1.0
Choline bitartrate 0.2
Cholesterol 0.25
BHT 0.02
The total fat content of the control diet was 5% fed in the form of a mix of lard and safflower oil to provide a P/S ratio of 0.4. The control diet alone will be fed to Group 1. For identity of diets in Groups 2-8, see Figure 10 (a). Biomass and polysaccharide was matrixed into the oil component of the diets. Food intake and body weight of individual animals was monitored regularly through the feeding period.
For diets listed as Quadro ground, biomass was homogenized in a Quadro FlO rotor homogenizer (Quadro Engineering Inc., Waterloo, Ontario, Canada). For ball milled samples: samples were cryogenically ball milled in a planetary ball mill (Retsch, PMlOO) at 10-80 g batch size. The powder was placed in a grinding bowl with eight to ten %-inch- diameter stainless steel balls. The sample was cooled with liquid nitrogen repeatedly. The material was milled at 400-550 rpm for 30 to 60 min. The final product was dried in a desiccator overnight.
After 30 days on diets, hamsters in each group will be anaethesized with halothane and blood samples were collected and stored at -800C for determination of total cholesterol, low density lipoprotein (LDL), and high density lipoprotein (HDL) subclass cholesterol and triglyceride (TG) levels. Measures of antioxidant status were also determined through TBARs assays. Lipid analysis
Plasma total cholesterol was measured using a VG Autoanalyzer in conjunction with commercial enzymatic kits and appropriate standards (Abbott Diagnostics, Montreal, Quebec). All samples were processed through the above system in duplicate.
The results are shown in Figure 10 (b) and (c), where each of Groups 2-8 exhibited a decrease in total serum cholesterol (TSC) levels in comparison to the control diet (Group 1). The lowering of total serum cholesterol levels ranged from about 35 to about 62%.
The results reflect an unexpected discovery compared to earlier reports. For example, Dvir et al. reported only a 21.8% reduction in TSC by feeding rodents microalgal biomass at 19% of the total diet. This is in contrast to the 5% of the total diet used in Group 3 and the 10% of the total diet in Group 5 of the instant experiment, which produced reductions of 35.9% and 54.2%, respectively. The results from the instant experiment are also unexpected relative to the report by Ginzberg et al., where 5% and 10% diets only produced 11% and 28% reductions in TSC, respectively. The results from Group 7 (62% reduction) also indicate the presence of unexpected synergy in a combination of cell homogenate and an additional agent such as phyto sterol.
EXAMPLE 20
The effect of disclosed compositions on anthropometric indices was studied in the hamsters described in Example 19.
Body weight was measured once a week and food consumption once every two days by weighing the difference of food cups before and after the feeding. Measurements of body composition were performed using DEXA analyses at sacrifice. Methods to determine body composition and energy expenditure (metabolism) were performed essentially as described by Lei et al. ("Relationship of total body fatness and five anthropometric indices in Chinese aged 20-40 years: different effects of age and gender" Eur. J. Clin. Nutri. (2006) 60:511-518) and Nagao et al. ("The lOtrans, 12 cis isomer of conjugated linoleic acid promotes energy metabolism in OLETF rats" Nutri. (2003) 19:652-656). Increasing satiety
The average food intake results are shown in Figure 11 (a), where some of Groups 2- 8 exhibited a decrease in average food intake in comparison to the control diet (Group 1). The results reflect an increased satiety, and so decreased appetite, seen in at least the hamsters of Groups 4 and 5. The effect on body weight is shown in Figure 1 l(b). While the invention is not limited by theory, it is believed that the propensity of the polysaccharide to swell in size on contact with water in the gut, thus signaling a feeling of satiety in mammals. Body composition
The body composition (total fat mass) results are shown in Figure 12, where each of Groups 2-8 exhibited a decrease in total body fat mass in comparison to the control diet (Group 1). The results reflect a decrease in body fat (or hypolipidemic effect) seen in the hamsters of Groups 2-8. Energy expenditure
The energy expenditure results are shown in Figure 13, where some of Groups 2-8 exhibited an increase in energy expenditure in comparison to the control diet (Group 1). The results reflect increased energy use (and oxygen consumption) in at least the hamsters of Groups 4, 6 and 7.
EXAMPLE 21 Antioxidant status
The effect of disclosed compositions on antioxidant status was studied in the hamsters described in Example 19. Measurement of TBARs in frozen plasma were conducted using standardized commercially available kits (Catalog #: 0801192, ZeptoMetrix Corporation, Buffalo, NY), against appropriate standards. The plasma samples were frozen after collection until used to determine oxidative status by measuring thiobarbituric acid-reactive substances (TBARs). These analyses were conducted in duplicate to reduce analytical variability associated with these measurements. The results are shown in Figure 14, where each of Groups 2-5, 7, and 8 displayed a decrease in TBARs in comparison to the control diet (Group 1). A comparison of the results for Groups 2 and 6, for example, indicate an unexpected discovery that homogenization or disruption of the cells (biomass), even when administered at the same dosage, results in an increase an antioxidant status in the plasma of the treated animals. Given an understanding of the microalgal polysaccharides as on the surface of the cells or extracellular, there was no expectation that rupturing the microalgal cells in a feed composition fed to animals would increase the antioxidants in the animal. However, the results clearly indicate the superiority of homogenized or disrupted biomass (cells).
These results demonstrate that compositions provided herein are useful, for example, in alleviating age-associated cognitive and motor changes in mammals. See for example Brain Res. 1995 Sep 25;693(l-2):88-94.
EXAMPLE 22
A Porphyridium sp. biomass homogenate was tested for COXl and COX2 inhibition activity. Porphyridium sp. biomass was grown as described in Example 1 , recovered by centrifugation, washed with deionizer water to remove salts, and centrifugd again to form a pelleted paste. Nitrogen was bubbled through the paste for 30 minutes to displace dissolved oxygen and minimize subsequent oxidation. The paste was then passed through a model 11 OY Microfluidics Microfluidizer® at 22,000 PSI with cooling, and the process repeated until at least 50% of the cells were broken as determined by microscopic examination. Nitrogen was once again bubbled through the paste, which wais then lyophilized after shell freezing in dry ice ethanol. The dried cell mass was then ground to a fine powder with a Braun® kitchen homogenizer.
The powder was analyzed for inhibition of constitutive cyclooxygenase (COXl) and inducible cyclooxygenase (COX2) at 370C by monitoring oxygen consumption using an Oxytherm Electrode Unit by Hansatech. The ICs0 values are equal to the concentration of the sample that inhibits 50% activity of the cyclooxygenase in the reaction mixture. The results are shown in Table 14.
Table 14
biomass powder
Results indicate surprisingly significant anti-inflammatory activity, particulary for a natural product.
Certain aspects of the invention are based upon the discovery of an antiinflammatory effect in animals that ingest the cell material (biomass, homogenate, and/or polysaccharides) described herein. While the basis of the anti-inflammatory effect has yet to be completely elucidated, the materials of the invention both increase the antioxidant status of mammalian plasma (see Example 23 below) and inhibit cyclooxygenase activity. The cell material has anti -inflammatory activity demonstrated through multiple methods of analysis and shown to be exerted through independent meachansism that would not have been expected.
Animal models that are widely viewed to reflect inflammatory responses and to have predictive value in assessing the efficacy of various treatments for these disorders can be utilized to evaluate the therapeutic efficacy of the compounds described herein. The effects of the compounds of the invention on joint inflammation and lameness can be assessed in horses with trauma-induced inflammation. Alternatively, improvement of inflammation and mobility can be measured with a collagen-induced rheumatoid arthritis mouse model (see, Enokida M et al. (2001) Bone 28(1): 87-93)
EXAMPLE 23
Two filter-purified polysaccharide preparations (containing 3.57% and 4.23% sulfur by weight), prepared as described in Example 24, were subjected to oxygen radical absorption capacity (ORAC) analysis as described in Journal of Agricultural and Food Chemistry.; 2001 ;49(10); 4619-4626 and Journal of Agricultural and Food Chemistry.; 2002, 50(7); 1815-1821 , The polysaccharide containing 3.57% sulfur by weight did not exhibit ORAC activity. The polysaccharide containing 4.23% sulfur by weight exhibited ORAC activity of 2.0 trolox equivalents per gram.
EXAMPLE 24 Porphyridium cruentum and Porphyridium sp. were grown in artificial seawater media essentially as described in Example 1 except that the amount OfMgSO4 was varied. Porphyridium sp.cells were grown in 17mM MgSO4. Porphyridium cruentum was grown in 12OmM, 60OmM, 750 mM, IM3 and 2M MgSO4. Cell division occurred at all concentrations. Polysaccharide was purified essentially as described in Example 2 from the 120 and 60OmM cultures, to the point where all soluble protein and small molecules were removed. Sulfur content was analyzed according to US EPA SW846, Method 6010B, Inductively Coupled Plasma- Atomic Emission Spectrometry. The polysaccharide purified from the 17, 120 and 60OmM cultures contained 3.57, 4.23 and 5.57% sulfur, respectively. As disclosed herein, these results reflect an unexpected discovery because the skilled person would not expect that the microalgae would survive in media containing more than 100 mM sulfate.
EXAMPLE 25
Transformation of Porphyridium and Genotyping
The Porphyridum glycoprotein promoter, SEQ ID NO:44, was cloned in operable linkage with a zeocin resistance ble cDNA containing small 5' and 3' flanking regions (SEQ ID NO: 70) encoding the protein of SEQ ID NO: 1 1, with the far 5' region of the glycoprotein promoter directly adjacent to the pBlusecript Sacl site and the far 3' region of the CMV 3'UTR (SEQ ID NO:69) adjacent to the pBlusecript Kpnl site. The CMV 3'UTR was also in operable linkage with the ble cDNA. The plasmid was linearized by Kpnl,. which does cut in the promoter, ble cDNA, or 3'UTR, prior to transformation.
The biolistic particle delivery system PDS 1000/ He (Bio-Rad, USA) was used for transformation. Porphyridium sp. culture was grown to logarithmic phase (~2 x 106 cells/mL) in liquid ATCC 1495 media under continuous light (approximately 75 umol/photons/m2). Cells from this culture were harvested at 4,000 rpm at room temperature. The cell pellet was washed twice with sterile distilled water. Cells were resuspended in fresh ATCC 1495 media to the same cell density i.e.. ~2 x 106 cells/mL and incubated in the dark for 16 hours. The dark adapted cells were then harvested at 4000 rpm at room temperature, resuspended in fresh ATCC 1495 media to a density of ~2 x 108 cells/mL. Approximately 1 x 10s cells were transferred to each ATCC 1495 agarose plate. Filter sterilized DNA from the plasmids was coated onto 550 nm gold particles (catalog number S04e, Seashell Technology, USA) according to the manufacturer's protocol. For each of the particle bombardments, 1 ug of plasmid DNA was used. The negative controls were bombarded in identical fashion with gold particles coated with a plasmid containing the Porphyridum glycoprotein promoter, SEQ ID NO:44, and the CMV 3'UTR, (SEQ ID NO:69), with no zeocin resistance gene. Each of the particle bombardments were performed using 1350 psi rupture disks, at bombardment distance of 9 cm, and under 28 in. Hg vacuum. The bombarded cells were scraped off the plates, and transferred to 100 ml of fresh ATCC 1495 media, and shaken under continuous light (approximately 75 umol/ m2) for 3 days. Following recovery, the cells were harvested at 4,000 rpm at room temperature, and plated onto ATCC 1495 plates supplemented with 30 ug/mL Zeocin (Invitrogen, Carlsbad, CA, USA) at a cell density of 1 x 107 cells/plate. These plates were incubated under light (approximately 25 umol/ m2) for 4-5 weeks. Zeocin resistant colonies growing on these plates were scored as transformants and transferred onto fresh ATCC 1495 plates supplemented with 30 ug/mL Zeocin (Invitrogen, Carlsbad, CA, USA) for growth and analysis.
Zeocin resistant colonies appeared after 2-3 weeks. Genotyping with primers specific to the zeocin resistance gene was performed on genomic DNA isolated from zeocin resistant colonies. Results from genotyping of one strain (referred to herein and labeled as "transformant #2" in Figure 15) indicated that the zeocin resistance gene was present. A band of the correct size was amplified. Results are shown in Figure 5 and discussed in more detail in Example 21.
EXAMPLE 26
Transformation of Porphyridium, Genotyping, and Southern Blot Analysis The zeocin resistance plasmid described in Example 20 and a second plasmid that was identical with the exception that it contained a cDNA for a human GLUTl glucose transporter (SEQ ID NO:25) instead of the ble cDNA were combined in a co-transformation experiment carried out essentially as described in Example 20 except that both the zeocin resistance and GLUTl plasmids were both adhered to the gold beads. A zeocin resistant colony (referred to herein as transformant#l) was selected for further analysis. Genomic DNA was extracted from wild type Porphyridium sp. and transformant#l.
Genotyping was performed on genomic DNA extracted from wild type, transformant#l, and transformant#2 DNA with plasmid DNA used as a template positive control and water in place of DNA as a template negative control. A segment of the Porphyridium glycoprotein (GLP) gene promoter was used as a target positive control. The following primer sets were used for the genotyping PCR: BIe- FWD (SEQ ID NO: 62) and BIe-REV (SEQ ID NO: 63), GLP-FWD (SEQ ID NO: 64) and (SEQ ID NO: 65), GLUTl- FWD (SEQ ID NO: 66) and GLUTl-REV (SEQ ID NO: 67). The PCR profile used was as follows: 94°C denaturation for 5 min; 35 cycles of 94°C for 30 sec, 510C or 6O0C (510C for glycoprotein gene & GLUTl and 6O0C for ble) for 30 sec, 72°G for 2 min; 720C for 5 min. Results are shown in Figure 15. Figure 15 (a) demonstrates that the ble gene was present in both transformants, as the expected 300bp product was generated. Figure 15(b) demonstrates that the genomic DNA extraction and amplification was working, as the expected 948bp glycoprotein promoter fragment was generated. Figure 15(c) demonstrates that the GLUTl gene was present transformant #1, as the expected 325bp product was generated. DNA ladder was from BioNexus, Inc., All Purpose Hi-Lo DNA Marker, Catalog No: BN2050.
Specific bands can be amplified from residual plasmid DNA adhered to the outside of cells on transformation plates. Additionally, plasmids that have not been linearized can be maintained as episomes for a period of time before being degraded and can serve as template during PCR despite not having been integrated into a chromosome of a host organism. In both cases, microalgal strains may genotype positive despite the absence of stable chromosomal integration of the vector. Antibiotic resistant strains are known to arise due to mutagenesis caused by chromosomal damage from biolistic particles, electroporation conditions, and random genetic variation that is known to occur in microbial organisms. Southern blot analysis was performed to conclusively confirm the integration of the GLUTl construct into the genome of transformant #1.
Southern blot analysis was performed on transformant #1. 20 μg genomic DNA from wild type and transformant#l were individually digested with Hinc II, Sac I , Xho I and separated on a 1% agarose gel. DNA was transferred onto Nylon membrane (Hybond N+, Amersham Biosciences). A 1495 bp fragment containing the entire coding region of the GLUTl gene was used as a probe. DIG labeled probes were generated for each probe fragment using the DIG High Prime DNA labeling and detection Kit according to the manufacturers instructions (Roche). The hybridizing bands were detected using the colorimetric detection reagents provided by the manufacturer. Figure 16 demonstrates that the GLUTl plasmid was stably integrated into the genome of transformant#l while no detectable signal arose from wild type genomic DNA. As would be expected for a plasmid integrating into a chromosome of an organism, the specific band was in a different position for each different restriction enzyme used to digest the genomic DNA. This data conclusively demonstrates a species of the genus Porphyridium containing an exogenous gene encoding a carbohydrate transporter integrated into a chromosome of the organism. In this embodiment the carbohydrate transporter gene is in operable linkage with a promoter endogenous to a species of the genus Porphyridium. In some other embodiments embodiment the carbohydrate transporter gene is in operable linkage with a promoter endogenous to a Rhodophyte.
EXAMPLE 27
Hyaluronidase inhibition
Biotinylated hyaluronic acid (bHA) was covalently attached to the wells of a 96-well plate. Samples containing hyaluronidase and various test materials were then added to the wells. The hyaluronidase degrades the bound hyaluronic acid, resulting in a decrease in the amount of biotin covalently linked to the well plate. At the end of the incubation period the reaction was stopped and the well plate was washed to remove the hyaluronidase. The remaining bHA was detected using an avidin bound peroxidase enzyme. When an appropriate substrate is added, the peroxidase enzyme generated a color signal in proportion to the amount of bHA. The color signal was measured spectrophotometrically, and was inversely proportional to the amount of hyaluronidase activity in the sample. Thus, materials that inhibited hyaluronidase resulted in a greater color signal, since more of the bHA remained intact. Also see Frost, G., L, Stern, R. A Microtiter-Based Assay for Hyaluronidase Activity Not Requiring Specialized Reagents. Analytical Biochemistry 251, 263-269: 1997.
Preparation of Test Material Extracts
Test Material A was supplied as a powder type material. For this study, 100 mg of this material was combined with either 5 ml of ethanol or 5 ml of ultrapure water in 15 ml centrifuge tubes. After combining, the mixtures were vortexed, then placed onto a rocking platform for approximately 30 minutes at room temperature, and then centrifuged at 1,000 RPM for 5 minutes. The supernatants were then used at the final concentrations listed in the results section. Test Material B was supplied as a thick, viscous solutions (3%).
Anti -Hyaluronidase Assay: Immobilization of bHA onto 96-well plates
A solution of sulfo-NHS (0.184 mg/ml) and bHA (0.2 mg/ml) was prepared in distilled water. 50 μl of this solution was then added to the wells of a 96-well Covalink-NH plate. A solution of EDC (0.123 mg/ml) was then prepared in distilled water and 50 μl of this solution was added to each well (which resulted in a final concentration of 10 μg/well bHA and 6.15 μg/well of EDC). The well plate was then incubated overnight at 4±2°C or for 2 hours at room temperature. After the incubation the plate was washed three times with PBS containing 2 M NaCl and 50 mM MgSO4.
Anti-Hyaluronidase Assay
Prior to the assay, the well plate was equilibrated with assay buffer (0.1 M formate [pH 4.5], 0.1 M NaCl, 1% Triton X-IOO3 5 mM saccharolactone). The test materials were prepared in assay buffer at 2x their final concentration (heparin was used as a positive control, 1 mg/ml final concentration). After removing the assay buffer from the well plate used for equilibration, 50 μl of each of the prepared test materials was added to three wells on the well plate, followed by the addition of 50 μl of assay buffer containing hyaluronidase will be added to each well (0.1 mg/ml). Three additional wells were treated with 100 μl of assay buffer alone (without test materials and without hyaluronidase) and served as an index of zero hyaluronidase activity. After the addition of the test materials and enzyme, the plate was incubated for 30 minutes at room temperature. At the end of the incubation period, 200 μl of 6 M guanidine-HCl was added to each well to terminate the reaction. The plate was then washed three times with PBS containing 2 M NaCl, 50 mM MgSO4 and 0.05% Tween 20.
During the 30 minute incubation, an avidin/biotin-peroxidase complex was prepared in 10.5 ml of PBS containing 0.1% Tween 20 using an ABC kit. This mixture was incubated for at least 30 minutes prior to use. After the plate was washed, 100 μl of the avidin/biotin-peroxidase solution was added to each well and the plate was incubated for another 30 minutes at room temperature. The plate was washed three times with PBS containing 2 M NaCl, 50 mM MgSO4 and 0.05% Tween 20. After the final wash, 100 μl of substrate solution (one 10 mg tablet of OPD in 10 ml of 0.1 M citrate-PO4 buffer supplemented with 7.5 μl of 30% H2O2) was added to each well. The plate was incubated in the dark for 10-20 minutes and then read at 460 ran using a plate reader. The substrate solution was also added to three wells that were not treated with test materials or the avidin/biotin-peroxidase solution and were used as a blank for the absorbance measurements.
Table 13 Treatment Percent Inhibition
10% A Water Extract 86
5% MATERIAL A Water Extract 67
1 % MATERIAL A Water Extract 29
1.5% MATERIAL B 93 0.5% MATERIAL B 81 0.1% MATERIAL B 70
0.1% Heparin 74 Negative Control 0
Test Material Identification: MATERIAL A: Porphyridium sp. biomass homogenized (Quadro FlO); cells grown as described in Example 1
Physical Description: Red/Purple powder Concentrations Tested: 10%, 5%, 1% (Extracted in either ethanol or water)
Test Material Identification: 3% MATERIAL B: Exopolysaccahride from Porphyridium sp. purified as described in Example 2
Physical Description: Light tan, viscous liquid Concentrations Tested: 1.5%, 1%, 0.5%, 0.1%
Table 15: Beta-carotene hydroxylases
Genbank accession number
NP_682690
YP_172377
NP_440788
YP_475340
YP_475340
BAB75708
ZP_01084736
ZP_01080915
ZPJ) 1 123496
ABB27016
NP_895643
NP_896386 •
ABB34062
YP_292794
AAP99312
ZP_01006041
ABB49299
NP_848964
ZP_01040435
NP_049051 γp_457405
AAT38625
Table 16: Superoxide dismutases
Genbank accession number
CAA42737 CAA43859 AAA29934 BAA02655
NP_625295
AH004200
CACl 4367
YPJ)01003721
ABM60577
CAM08755
YP_966270
YP_963716
ABM37237
ABM35060
ABM33234
ABM33141
NP_407488
NP_405922
YP_932026
ZPJ) 1641300
ZP_01638301
ZP_01637188
EAX24391
EAX23794
EAX23720
EAX23627
EAX20859
EAXl 9390
EAXl 6596
CAL93139
YP_914326
YPJ747424
ABI59459
ZPJ)1610569
ZPJ) 1605216
ZPJ) 1600343
ZP_01584712
ZPJ)1581863
ZPJ)1581157
ZPJ) 1575777
ZPJ)1569848
ZPJ) 1559998
EAWO 1367
EAWO 1065
EAV97274
EAV95856
EAV80568
EAV73624
EAV73531
EAV70130
EAV66456
EAV61854
ZPJ)1532079 ZP_01516088
EAV26209
YP_845889
YP_822355
YP_843115
YP_836186
ABKl 7454
All references cited herein, including patents, patent applications, and publications, are hereby incorporated by reference in their entireties, whether previously specifically incorporated or not.
Having now fully described this invention, it will be appreciated by those skilled in the art that the same can be performed within a wide range of equivalent parameters, concentrations, and conditions without departing from the spirit and scope of the invention and without undue experimentation.
While this invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications. This application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains and as may be applied to the essential features hereinbefore set forth.

Claims

WHAT IS CLAIMED IS:
1. A composition comprising a homogenate of red microalgae cells.
2. The method of claim 1 , further comprising a carrier suitable for human consumption.
3. The composition of claim 2, wherein the red microalgae cells are of the genus Porphyridium.
4. The composition of claim 3, wherein the red microalgae cells are Porphyridium sp. UTEX 637 or a strain derived from Porphyridium sp. UTEX 637.
5. The composition of claim 3, wherein the red microalgae cells are Porphyridium omentum UTEX 161 or a strain derived from Porphyridium omentum UTEX 161.
6. The composition of claim 3, wherein the red microalgae cells are Porphyridium aerugineum or a strain derived from Porphyridium aerugineum.
7. The composition of claim 3, wherein the red microalgae cells are Porphyridium sordidum or a strain derived from Porphyridium sordidum.
8. The composition of claim 3, wherein the red microalgae cells are Porphyridium purpureum or a strain derived from Porphyridium purpureum.
9. The composition of claim 1, wherein the homogenate contains a sulfated exopolysaccharide secreted by the red microalgae cells that contains at least 4.6% sulfur by weight.
10. The composition of claim 9, wherein the homogenate contains a sulfated exopolysaccharide secreted by the red microalgae cells that contains at least 4.75% sulfur by weight.
11. The composition of claim 10, wherein the homogenate contains a sulfated exopolysaccharide secreted by the red microalgae cells that contains at least 5.0% sulfur by weight.
139
12. The composition of claim 11 , wherein the homogenate contains a sulfated exopolysaccharide secreted by the red microalgae cells that contains at least 5.5% sulfur by weight.
13. The composition of claim 12, wherein the homogenate contains a sulfated exopolysaccharide secreted by the red microalgae cells that contains at least 6.0% sulfur by weight.
14. The composition of claim 13, wherein the homogenate contains a sulfated exopolysaccharide secreted by the red microalgae cells that contains at least 7.0% sulfur by weight.
15. The composition of claim 1 , wherein the microalgal cell homogenate contains at least two times the amount of solvent-available polysaccharide present in a quantity of unhomogenized cells needed to generate the microalgal cell homogenate.
16. The composition of claim 15, wherein the microalgal cell homogenate contains at least three times the amount of solvent-available polysaccharide present in a quantity of unhomogenized cells needed to generate the microalgal cell homogenate.
17. The composition of claim 16, wherein the microalgal cell homogenate contains at least five times the amount of solvent-available polysaccharide present in a quantity of unhomogenized cells needed to generate the microalgal cell homogenate.
18. The composition of claim 17, wherein the microalgal cell homogenate contains at least ten times the amount of solvent-available polysaccharide present in a quantity of unhomogenized cells needed to generate the microalgal cell homogenate.
19. The composition of claim 18, wherein the microalgal cell homogenate contains at least twenty times the amount of solvent-available polysaccharide present in a quantity of unhomogenized cells needed to generate the microalgal cell homogenate.
20. The composition of claim 1 , further comprising a plant phytosterol.
140
21. The composition of claim 20, wherein the weight/weight ratio of homogenate to plant phytosterol is at least 2: 1.
22. The composition of claim 21 , wherein the weight/weight ratio of homogenate to plant phytosterol is at least 20:1.
23. The composition of claim 22, wherein the weight/weight ratio of homogenate to plant phytosterol is least 100: 1.
24. The composition of claim 20, wherein the plant phytosterol is a preparation of wood-derived phytosterol.
25. The composition of claim 24, wherein the wood-derived phytosterol preparation comprises at least 8% stanol, at least 40% sitosterol, and at least 20% campesterol.
26. The composition of claim 24, wherein the wood-derived phytosterol preparation comprises between 5% and 20% stanol, between 40% and 60% sitosterol, and between 15% and 35% campesterol.
27. The composition of claim I5 further comprising a limonoid.
28. The composition of claim 27, wherein the limonoid is selected from the group consisting of limonin and nomilin.
29. The composition of claim 27, wherein the weight/weight ratio of homogenate to limonoid is between 900: 1 and 25:1.
30. The composition of claim 1, further comprising a flavonoid.
31. The composition of claim 30, wherein the flavonoid is selected from the group consisting of hesperidin, naringin, naringenin, hesperitin, nobiletin and tangeretin.
32. The composition of claim 30, wherein the weight/weight ratio of homogenized red microalgae cells to flavonoid is between 800: 1 and 40: 1.
33. The composition of claim 1, further comprising a tocotrienol.
141
34. The composition of claim 33, wherein the tocotrienol is selected from the group consisting of an alpha- tocotrienol, gamma tocotrienol., and delta-tocotrienol.
35. The composition of claim 33, wherein the weight/weight ratio of homogenized red microalgae cells to tocotrienol is between 900:1 and 30: 1.
36. The composition of claim 1, further comprising at least two compounds selected from the group consisting of a plant phytosterol, a limonoid, a flavonoid, and a tocotrienol.
37. The composition of claim 36, wherein the composition comprises a plant phytosterol and at least one flavonoid selected from the group consisting of hesperidin, naringin, naringenin, hesperitin, nobiletin and tangeretin.
38. The composition of claim 20, wherein the homogenate contains a sulfated exopolysaccharide secreted by the red microalgae cells that contains at least 3.0% sulfur by weight; and the homogenate contains at least two times the amount of solvent-available polysaccharide present in a quantity of urihomogenized cells needed to generate the microalgal cell homogenate.
39. The composition of claim 27, 30 or 33, wherein a. the homogenate contains a sulfated exopolysaccharide secreted by the red microalgae cells that contains at least 4.75% sulfur by weight; and b. the homogenate contains at least two times the amount of solvent- available polysaccharide present in a quantity of unhomogenized cells needed to generate the microalgal cell homogenate.
40. The composition of claim 1, wherein the composition is a food.
41. The composition of claim 1, wherein the composition is a tablet.
42. The composition of claim 1, wherein the composition is a capsule.
43. A method of lowering cholesterol in a patient comprising orally administering a composition comprising a homogenate of red microalgae cells to a patient and thereby lowering the serum cholesterol.
142
44. The method of claim 43, wherein the composition further comprises a carrier suitable for human consumption.
45. The method of claim 43, wherein the cells are of the genus Porphyridium.
46. The method of claim 45, wherein the cells are Porphyridium sp. UTEX 637 or a strain derived from Porphyridium sp. UTEX 637.
47. The method of claim 45, wherein the cells are Porphyridium cruentum UTEX 161 or a strain derived from Porphyridium cruentum UTEX 161.
48. The method of claim 45, wherein the cells are Porphyridium aerugineum or a strain derived from Porphyridium aerugineum.
49. The method of claim 45, wherein the cells are Porphyridium sordidum or a strain derived from Porphyridium sordidum.
50. The method of claim 45, wherein the cells are Porphyridium purpureum or a strain derived from Porphyridium purpureum.
51. The method of claim 43, wherein the homogenate contains a sulfated exopolysaccharide secreted by the red microalgae cells that contains at least 4.6% sulfur by weight.
52. The method of claim 51, wherein the homogenate contains a sulfated exopolysaccharide secreted by the red microalgae cells that contains at least 4.75% sulfur by weight.
53. The method of claim 52, wherein the homogenate contains a sulfated exopolysaccharide secreted by the red microalgae cells that contains at least 5.0% sulfur by weight.
54. The method of claim 53, wherein the homogenate contains a sulfated exopolysaccharide secreted by the red microalgae cells that contains at least 5.5% sulfur by weight.
143
55. The method of claim 54, wherein the homo genate contains a sulfated exopolysaccharide secreted by the red microalgae cells that contains at least 6.0% sulfur by weight.
56. The method of claim 55, wherein the homogenate contains a sulfated exopolysaccharide secreted by the red microalgae cells that contains at least 7.0% sulfur by weight.
57. The method of claim 43, wherein the homogenate contains at least two times the amount of solvent-available polysaccharide present in a quantity of unhomogenized cells needed to generate the microalgal cell homogenate.
58. The method of claim 57, wherein the homogenate contains at least three times the amount of solvent-available polysaccharide present in a quantity of unhomogenized cells needed to generate the microalgal cell homogenate.
59. The method of claim 58, wherein the homogenate contains at least five times the amount of solvent-available polysaccharide present in a quantity of unhomogenized cells needed to generate the microalgal cell homogenate.
' 60. The method of claim 59, wherein the homogenate contains at least ten times the amount of solvent-available polysaccharide present in a quantity of unhomogenized cells needed to generate the microalgal cell homogenate.
61. The method of claim 60, wherein the homogenate contains at least twenty times the amount of solvent-available polysaccharide present in a quantity of unhomogenized cells needed to generate the microalgal cell homogenate.
62. The method of claim 43, wherein the composition further comprises a plant phytosterol.
63. The method of claim 62, wherein the weight/weight ratio of homogenate to plant phytosterol is at least 2:1.
64. The method of claim 63, wherein the weight/weight ratio of homogenate to plant phytosterol is at least 20: 1.
144
65. The method of claim 64, wherein the weight/weight ratio of homogenate to plant phytosterol is least 100: 1.
66. The method of claim 62, wherein the plant phytosterol is a preparation of wood-derived phytosterol.
67. The method of claim 66, wherein the wood-derived phytosterol preparation comprises at least 8% stanol, at least 40% sitosterol, and at least 20% campesteroi.
68. The method of claim 66, wherein the wood-derived phytosterol preparation comprises between 5% and 20% stanol, between 40% and 60% sitosterol, and between 15% and 35% campesterol.
69. The method of claim 43, wherein the composition further comprises a limonoid.
70. The method of claim 69, wherein the selected from the group consisting of limonin and nomilin.
71. The method of claim 69, wherein the weight/weight ratio of homogenate to limonoid is between 900: 1 and 25: 1.
72. The method of claim 43, wherein the composition further comprises a flavonoid.
73. The method of claim 72, wherein the flavonoid is selected from the group consisting of hesperidin, naringin, naringenin, hesperitin, nobiletin and tangeretin.
74. The method of claim 72, wherein the weight/weight ratio of homogenate to flavonoid is between 800: 1 and 40: 1.
75. The method of claim 1, wherein the composition further comprises a tocotrienol.
76. The method of claim 75, wherein the tocotrienol is selected from the group consisting of an alpha-tocotrienol, gamma tocotrienol, and delta-tocotrienol.
145
77. The method of claim 75, wherein the weight/weight ratio of homogenate to tocotrienol is between 900: 1 and 30: 1.
78. The method of claim 43, wherein the composition further comprises at least two compounds selected from the group consisting of a plant phytosterol, a limonoid, a flavonoid, and a tocotrienol.
79. The method of claim 78, wherein the composition comprises a plant phytosterol and at least one flavonoid selected from the group consisting of hesperidin, naringin, naringenin, hesperitin, nobiletin and tangeretin.
80. The method of claim previous 62, wherein: the homogenate contains a sulfated exopolysaccharide secreted by the red microalgae cells that contains at least 3.0% sulfur by weight; and the homogenate contains at least two times the amount of solvent-available polysaccharide present in a quantity of unhomogenized cells needed to generate the microalgal cell homogenate.
81. The method of claim previous 69, 72 and 75, wherein: the homogenate contains a sulfated exopolysaccharide secreted by the red microalgae cells that contains at least 3.0% sulfur by weight; and the homogenate contains at least two times the amount of solvent-available polysaccharide present in a quantity of unhomogenized cells needed to generate the microalgal cell homogenate.
82. The method of claim 43, wherein the composition is a food.
83. The method of claim 43, wherein the composition is a tablet.
84. The method of claim 43, wherein the composition is a capsule.
85. The method of claim 69, wherein the limonoid is administered is in the range of 1 to 500 mg/day.
86. The method of claim 72, wherein the flavonoid is administered is in the range of 200 to 5000 mg/day
146
87. The method of claim 75, wherein the tocotrienol is administered is in the range of 1 to 1200 mg/day
88. The method of claim 43, wherein the homogenate administered in the range of 0.005 to 5 grams per kilogram of mammalian body weight per day.
89. A composition comprising: a. a first component comprising cell material of the genus Porphyridium; and b. at least one additional component selected from the group consisting of: i. a plant phytosterol; ii. a limonoid; iii. a flavonoid; and iv. a tocotrienol; wherein the first and at least one additional component are packaged for sale as a single unit.
90. The composition of claim 89, wherein the first and at least one additional component are formulated in contact with each other.
91. The composition of claim 89, wherein the first and at least one additional component are formulated not in contact with each other.
92. The composition of claim 89, further comprising an instruction for mixing the first and at least one additional component in a specific ratio.
93. The composition of claim 89, further comprising instructions for ingesting the first and at least one additional component in specific dosages.
94. The composition of claim 89, further comprising instructions for lowering cholesterol.
95. The composition of claim 89, wherein the at least one additional component is a plant phytosterol is a preparation of wood-derived phytosterol.
147
96. The composition of claim 95, wherein the wood-derived phytosterol preparation comprises at least 8% stanol, at least 40% sitosterol, and at least 20% campesterol.
97. The composition of claim 95, wherein the wood-derived phytosterol preparation comprises between 5% and 20% stanol, between 40% and 60% sitosterol, and between 15% and 35% campesterol.
98. The composition of claim 89, wherein the at least one additional component is a limonoid selected from the group consisting of limonin and nomilin.
99. The composition of claim 89, wherein the at least one additional component is a flavonoid is selected from the group consisting of hesperidin, naringin, naringenin, hesperitin, nobiletin and tangeretin.
100. The composition of claim 89, wherein the at least one additional component is a tocotrienol is selected from the group consisting of an alpha-tocotrienol, gamma tocotrienol, and delta-tocotrienol.
101. A method of formulating a composition comprising: a. culturing cells of the genus Porphyridium; b. separating the cells from culture media; c. drying the cells; and d. mixing the cells with at least one compound selected from the group consisting of: i. a plant phytosterol ii. a limonoid; iii. a flavonoid; and iv. a tocotrienol.
102. The method of claim 101, wherein the cells are homogenized before mixing with the at least one compound to generate a homogenate.
103. The method of claim 102, wherein the cells are homogenized such that the homogenate contains at least two times the amount of solvent-available
148 polysaccharide present in a quantity of unhomogenized cells needed to generate the homogenate.
104. The method of claim 101, wherein the cells are cultured in at least 10O mM sulfate.
105. The method of claim 104, wherein the cells are cultured in at least 200 mM sulfate
106. The method of claim 105, wherein the cells are cultured in at least 300 mM sulfate.
107. The method of claim 106, wherein the cells are cultured in at least 500 mM sulfate.
108. The method of claim 107, wherein the cells are cultured in at least 600 mM sulfate.
109. The method of claim 108, wherein the cells are cultured in at least 700 mM sulfate.
110. The method of claim 102, wherein the cells are washed before homogenization.
111. The method of claim 102, wherein the homogenate contains a sulfated exopolysaccharide secreted by the Porphyridium cells that contains at least 4.6% sulfur by weight.
112. The method of claim 111, wherein the homogenate contains a sulfated exopolysaccharide secreted by the Porphyridium cells that contains at least 4.75% sulfur by weight.
113. The method of claim 112, wherein the homogenate contains a sulfated exopolysaccharide secreted by the Porphyridium cells that contains at least 5% sulfur by weight.
149
114. The method of claim 113, wherein the homogenate contains a sulfated exopolysaccharide secreted by the Porphyridium cells that contains at least 5.5% sulfur by weight.
115. The method of claim 114, wherein the homogenate contains a sulfated exopolysaccharide secreted by the Porphyridium cells that contains at least 6% sulfur by weight.
116. The method of claim 115, wherein the homogenate contains a sulfated exopolysaccharide secreted by the Porphyridium cells that contains at least 7% sulfur by weight.
117. The method of claim 116, further comprising the step of adding a carrier suitable for human consumption.
118. The method of claim 102, wherein the homogenization is performed by a method selected from the group consisting of pressure disruption, sonication, jet milling and ball milling.
119. The method of claim 104, wherein the sulfate is provided in at least one form selected from the group consisting OfNa2SO4-IO H2O, MgSθ4-7H20, MnSO4, and CuSO4.
120. The method of claim 101, further comprising the step of adding a carrier suitable for oral consumption.
121. The method of claim 101, wherein the drying is performed by tray drying, spin drying, rotary drying, spin flash drying, or lyophilization.
122. A method of reducing reactive oxygen species in mammals comprising administering the composition of any of claims 1 -42.
123. A method of delaying the onset of or treating a neurodegenerative disease comprising administering the composition of any of claims 1 -42.
150
124. The method of claim 123 wherein the disease is selected from the group consisting of Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, Freidreich's ataxia, Huntington's disease, and a Prion disease.
125. A method of reducing organ transplant rejection comprising administering the composition of any of claims 1-42.
126. A method of reducing the percentage of a mammalian body that is made of fat comprising administering the composition of any of claims 1 -42.
127. A method of increasing satiety comprising administering the composition of any of claims 1-42.
128. A method of increasing the energy expenditure of a mammal comprising administering the composition of any of claims 1 -42.
129. A method of reducing the body weight of a mammal comprising administering the composition of any of claims 1 -42.
130. A method of reducing inflammation in a mammal comprising administering the composition of any of claims 1-42.
131. A method for the prevention and/or treatment of atherosclerosis comprising administering the composition of any of claims 1 -42. 132. A cell of the genus Porphyridum, wherein the genome of the cell contains an exogenous gene encoding a carbohydrate transport protein. 133. The cell of claim 132, wherein the cell is of the species is selected from the group consisting of Porphyridium sp. UTEX 637 or a strain derived from Porphyridium sp. UTEX 637, Porphyridium cruentum UTEX 161 or a strain derived from Porphyridium cruentum UTEX 161, Porphyridium aerugineum or a strain derived from Porphyridium aerugineum, Porphyridium sordidum or a strain derived from Porphyridium sordidum, Porphyridium purpureum or a strain derived from Porphyridium purpureum. 134. The cell of claim 133, wherein the cell is of the species Porphyridium sp. UTEX 637 or a strain derived from Porphyridium sp. UTEX 637.
151
135. The cell of claim 132, wherein the carbohydrate transport protein has at least 60% amino acid identity with a member of the group consisting of SEQ ID NOs: 20, 22, 24, 26, 27, 29-39, and 46-48. 136. A composition comprising: a. polysaccharide isolated from a cell of the genus Porphyridium; and b. hyaluronic acid. 137. The composition of claim 136, wherein the composition is substantially free of endotoxins. 138. The composition of claim 136 wherein the composition is completely free of endotoxins. 139. The composition of claim 136 wherein the composition is substantially free of protein. 140. The composition of claim 136 wherein the composition is completely free of protein. 141. The composition of claim 136 wherein the cell is selected from the group consisting of Porphyridium sp. UTEX 637 or a strain derived from Porphyridium sp. UTEX 637, Porphyridium cruentum UTEX 161 or a strain derived from Porphyridium cruentum UTEX 161, Porphyridium aerugineum or a strain derived from Porphyridium aerugineum, Porphyridium sordidum or a strain derived from Porphyridium sordidum, and Porphyridium purpureum or a strain derived from Porphyridium purpureum. 142. The composition of claim 136, wherein the weight:weight ratio of hyaluronic acid to polysaccharide isolated from a cell of the genus Porphyridium is selected from the group consisting of at least 10,000:1, at least 1,000:1, at least 500:1, at least 250:1 , at least 125:1, at least 100:1, at least 50:1, at least 10:1, at least 5:1, at least 4:1, at least 3:1, at least 2:1; at least 1.5:1, approximately 1 :1, at most 1 :10,000, at most 1:1,000, at most 1:500, at most 1:250, at most 1: 125:1, at most 1:100, at most 1 :50:1, at most 1 :10, at most 1 :5, at most 1 :4, at most 1 :3, at most 1 :2; and at most 1 :1.5. 143. The composition of claim 136, wherein the composition is sterile. 144. The composition of claim 136, wherein the composition further comprises a carrier suitable for parenteral administration. 145. The composition of claim 136, wherein the composition further comprises a carrier suitable for sub-dermal administration.
152
146. The composition of claim 136, wherein the composition further comprises a carrier suitable for dermal administration. 147. The composition of claim 136 wherein the polysaccharide isolated from a cell of the genus Porphyridium contains an amount of sulfur by weight selected from the group consisting of at least 3%, at least 3.5%, at least 4.0%, at least 4.5%, at least 5.0%, at least 5.5%, at least 6.0%, at least 6.5%, at least 7.0%, at least 7.5% and at least 8.0%. 148. A method of extending the half life of a composition comprising hyaluronic acid comprising co-administering polysaccharide isolated from a cell of the genus Porphyridium. 149. The method of claim 148, wherein the hyaluronic acid and polysaccharide isolated from a cell of the genus Porphyridium are in direct contact before the co-administration. 150. The method of claim 148, wherein the hyaluronic acid and polysaccharide isolated from a cell of the genus Porphyridium are not in direct contact before the co-administration. 151. The method of claim 148, wherein the hyaluronic acid and polysaccharide isolated from a cell of the genus Porphyridium are administered separately and come into direct contact only after both are inside a mammalian tissue. 152. The method of claim 148, wherein the composition is injected into a mammalian joint. 153. The method of claim 152, wherein the joint is selected from the group consisting of fibrous, cartilaginous, and synovial. 154. The method of claim 152, wherein the joint is selected from the group consisting of knee, shoulder, elbow, hip, ankle, wrist, and vertebrae. 155. The method of claim 148, wherein the composition is injected into skin. 156. .The method of claim 155, wherein the skin is on a human face. 157. A method of zeaxanthin production comprising expressing an exogenous nucleic acid, wherein the nucleic acid encodes a protein that catalyzes the conversion of beta carotene to zeaxanthin in a microalgae cell that endogenously produces beta carotene. 158. The method of claim 157, wherein the protein has at least 25% amino acid identity with any protein listed in Table 4. 159. The method of claim 157, wherein the cell is selected from Table 1.
153
80 160. The method of claim 159, wherein the cell is of the genus
81 Porphyridium.
82 161. The method of claim 157, wherein the exogenous nucleic acid is
83 integrated into the nuclear genome of the cell and encodes a chloroplast targeting signal.
84 162. The method of claim 157, wherein the exogenous nucleic acid is
85 integrated in the chloroplast genome of the cell.
86 163. The method of claim 157, further comprising expressing an
87 exogenous nucleic acid sequence in the cell that encodes a protein that catalyzes the
88 transport of an exogenously provided fixed carbon source into the cell.
89 164. The method of claim 163, wherein the fixed carbon source is selected
90 from Table 2.
91 165. The method of claim 164, wherein the fixed carbon source is glucose.
92 166. A method of increasing the amount of zeaxanthin produced in
93 microalgae cells comprising:
94 a. transforming at least one first microalgae strain with an expression
95 vector encoding an enzyme that catalyzes the conversion of beta carotene to zeaxanthin to
96 produce a second strain; and
97 b. screening the second strain for production of more zeaxanthin per
98 unit volume of culture per unit time than the first strain.
99 167. The method of claim 166, wherein the microalgae strain is of the
100 genus Porphyridium.
101 168. The method of claim 166, wherein the first microalgae strain contains
102 an exogenous gene encoding an enzyme that catalyzes the conversion of lycopene to beta
103 carotene.
104 169. The method of claim 166, wherein the first microalgae strain contains
105 an exogenous gene encoding a protein that catalyzes the transport of an exogenously
106 provided fixed carbon source into the cell.
107 170. The method of claim 166, wherein the first microalgae strain
108 synthesizes less polysaccharide than strains UTEX 161 or UTEX 637.
109 171. A method of increasing the amount of zeaxanthin produced in
110 microalgae cells comprising:
111 a. transforming at least one first microalgae strain with an expression
112 vector encoding an enzyme that catalyzes the conversion of lycopene to beta carotene to
113 produce a second strain; and
154
.14 b. screening the second strain for production of more zeaxanthin per
L 15 unit volume of culture per unit time than the first strain.
L 16 172. The method of claim 171, wherein the microalgae strain is of the
117 genus Porphyridium.
L 18 113. The method of claim 171, wherein the first microalgae strain contains
1 19 an exogenous gene encoding an enzyme that catalyzes the conversion of beta carotene to
120 zeaxanthin.
121 174. The method of claim 171, wherein the first microalgae strain contains
122 an exogenous gene encoding a protein that catalyzes the transport of an exogenously
123 provided fixed carbon source into the cell.
124 175. The method of claim 171, wherein the first microalgae strain
125 synthesizes less polysaccharide than strains UTEX 161 or UTEX 637.
126 176. A method of increasing the amount of zeaxanthin produced in
127 microalgae cells comprising:
128 a. transforming at least one first microalgae strain with an expression
129 vector encoding a protein that catalyzes the transport of an exogenously provided fixed
130 carbon source into the cell to produce a second strain; and
131 b. screening the second strain for production of more zeaxanthin per
132 unit volume of culture per unit time than the first strain.
133 177. An expression vector comprising:
134 a. a segment of at least 25 nucleotides of SEQ ID NO: 57, and
135 b. a nucleic acid sequence that encodes a protein that
136 L has at least 25% amino acid identity with at least one sequence from
137 Table 15: and
138 ii. catalyzes the conversion of beta carotene to zeaxanthin.
139 178. The expression vector of claim 177, wherein the nucleic acid segment
140 of at least 25 nucleotides of SEQ ID NO:57 comprises the sub-sequence of SEQ ID NO:61.
141 179. A population of cells of the genus Porphyridium, wherein each cell
142 contains:
143 a. an exogenous nucleic acid that encodes a protein that catalyzes the
144 conversion of beta carotene to zeaxanthin; and
145 b. a ratio of zeaxanthin to beta carotene of at least 1.8:1.
146 180. The population of claim 179, wherein the protein has at least 25%
147 amino acid identity with any sequence from Table 4.
155
181. The population of claim 179, wherein each cell further contains an exogenous nucleic acid sequence encoding a protein that catalyzes the transport of an exogenously provided fixed carbon source into the cell. 182. The population of claim 179, wherein the ratio of zeaxanthin to beta carotene is at least 1.9: 1. 183. The population of claim 182, wherein the ratio of zeaxanthin to beta carotene is at least 2:1. 184. The population of claim 183 , wherein the ratio of zeaxanthin to beta carotene is at least 5:1. 185. The population of claim 184, wherein the ratio of zeaxanthin to beta carotene is at least 10:1. 186. The population of claim 185, wherein the ratio of zeaxanthin to beta carotene is at least 20: 1. 187. The population of claim 179, wherein each cell further contains an exogenous nucleic acid sequence encoding an enzyme that catalyzes the conversion of lycopene to beta carotene. 188. A plasmid containing: a. a segment of at least 20 nucleotides that is identical to one or more segments found in the nuclear or chloroplast genome of a cell of the genus Porphyridium; and b. a segment encoding a protein that converts one carotenoid into another distinct carotenoid; wherein the segment encoding a protein that converts one carotenoid into another distinct carotenoid is in operable linkage with a promoter active in microalgae. 189. The plasmid of claim 188, wherein the protein converts beta carotene into zeaxanthin. 190. The plasmid of claim 189, wherein the protein converts lycopene to beta carotene. 191. The plasmid of claim 188, wherein the segment of at least 20 nucleotides is identical to any segment from SEQ ID NO: 44. 192. The plasmid of claim 188, wherein the segment encoding a protein that converts one carotenoid into another distinct carotenoid contains only codons preferred in the chloroplast genome of a species of the genus Porphyridium.
156
81 193. The plasmid of claim 192, wherein the segment encoding a protein
82 that converts one carotenoid into another distinct carotenoid or xanthophyll contains only .83 codons most preferred in the chloroplast genome of a species of the genus Porphyridium. 184 194. A hydrophobic extract from cells of the genus Porphyridium, wherein 185 the extract contains :
186 a. EPA;
187 b. ARA; and
188 c. zeaxanthin.
189 195. The extract of claim 194, wherein the extract further comprises beta
190 carotene, and the ratio of zeaxanthin to beta carotene is at least 1.8:1.
191 196. The extract of claim 195, wherein the ratio of zeaxanthin to beta
192 carotene is at least 1.9 : 1.
193 197. The extract of claim 196, wherein the ratio of zeaxanthin to beta
194 carotene is at least 2:1.
195 198. The extract of claim 197, wherein the ratio of zeaxanthin to beta
196 carotene is at least 5:1.
197 199. The extract of claim 198, wherein the ratio of zeaxanthin to beta
198 carotene is at least 10:1.
199 200. The extract of claim 199, wherein the ratio of zeaxanthin to beta
200 carotene is at least 20: 1.
201 201. A method of producing zeaxanthin comprising culturing at least one
202 cell of the genus Porphyridium in the presence of a fixed carbon source in the absence of
203 light.
204 202. The method of claim 201 , wherein the fixed carbon source is selected
205 from the group consisting of glycerol, glucose and floridoside.
206 203. A method of increasing the amount of a carotenoid produced in
207 microalgae cells comprising:
208 a. treating the cells with a mutagen; and
209 b. screening mutagenized cells for production of more of the carotenoid per
210 unit volume of culture per unit time than unmutagenized cells.
21 1 204. The method of claim 203 , wherein the cells are of the genus
212 Porphyridium.
213 205. The method of claim 203, wherein the carotenoid is listed in Table 3.
214 206. The method of claim 205, wherein the carotenoid is zeaxanthin.
157 ! 15 207. A method of zeaxanthin production comprising:
116 a. culturing cells of the genus Porphyridium:
» 17 i. in the absence of light; and
118 ii. in the presence of an exogenously provided fixed carbon
.19 source; and
120 b. harvesting cells.
.21 208. The method of claim 207, further comprising extracting a
222 hydrophobic fraction from the harvested cells.
223 209. The method of claim 207, wherein the fixed carbon source is
224 glycerol.
225 ' 210. The method of claim 207, wherein the fixed carbon source is glucose.
226 21 1. The method of claim 207, wherein the fixed carbon source is
227 floridoside.
228 212. A method of treating an ocular disease comprising providing a
229 composition comprising cells of the genus Porphyridium, wherein the ratio of zeaxanthin to
230 beta carotene is at least 1.8:1, wherein the cells are formulated with a carrier suitable for
231 human consumption.
232 213. A method of producing an exopolysaccharide comprising:
233 a. culturing microalgae in a bioreactor, wherein gas is infused into
234 the bioreactor;
235 b. separating the microalgae from culture media, wherein the culture
236 media contains the exopolysaccharide; and
237 c. separating the exopolysaccharide from other molecules present in 238 the culture media.
239 214. The metho d of claim 213, wherein the microalgae is selected from
240 Table 1.
241 215. The method of claim 214, where the microalgae is of the genus
242 selected from the group consisting of Porphyridium, Rhodella, Chlorella, and Achnanthes.
243 216. The method of claim 215, wherein the microalgae is selected from
244 the group consisting of Porphyridium sp., Porphyridium omentum, Porphyridium
245 purpureum, Porphyridium aerugineum, Rhodella maculata, Rhodella reticulata, Chlorella
246 autotrophica, Chlorella stigmatophora, Chlorella capsulata, Achnanthes brevipes and
247 Achnanthes longipes.
158
.48 217. The method of claim 213, wherein the culture media contains at least
:49 HO mM sulfate.
150 218. The method of claim 217, wherein the sulfate is provided in at least
:51 one form selected from the group consisting of NazSCVlO H2O, MgSO4-VH2O, MnSO4, and
152 CuSO4.
!53 219. The method of claim 213, wherein the exopolysaccharide is separated
.54 from other molecules present in the culture media by tangential flow filtration.
Ϊ55 220. The method of claim 219, wherein a filter used for tangential flow
£56 filtration has a pore size from about 300Kd to about 0.45 micrometers.
257 221. The method of claim 213, wherein the exopolysaccharide is separated
258 from other molecules present in the culture media by alcohol precipitation.
259 222. The method of claim 221, wherein the alcohol is selected from the
260 group consisting of ethanol, isopropanol, and methanol.
261 223. The method of claim 213, further comprising the step of treating the
262 exopolysaccharide with a protease.
263 224. The method of claim 223, wherein the separating step comprises
264 separating the exopolysaccharide from proteins, peptides, and amino acids after protease
265 treatment.
266 225. The method of claim 213, wherein the bioreactor is a photobioreactor
267 and light is directed to a surface of the photobioreactor.
268 226. The method of claim 225, wherein aqueous culture medium is not
269 flowed through the photobioreactor after inoculation.
270 227. The method of claim 213, wherein the microalgae is cultured in the
271 presence of an exogenously provided fixed carbon source.
272 228. The method of claim 227, wherein the fixed carbon source is selected
273 from Table 2 or Table 3.
274 229. The method of claim 228, wherein the fixed carbon source is selected
275 from the group consisting of glucose, galactose, xylose, mannose, rhamnose, N-
276 acetylglucosamine, glycerol, floridoside, and glucuronic acid.
277 230. The method of claim 229, wherein the fixed carbon source is between
278 approximately 0.25% and approximately 0.75% glycerol vol/vol and the cells are cultured
279 in the presence of light.
280 231. The method of claim 230, wherein the fixed carbon source is
281 approximately 0.5% glycerol vol/vol.
159
82 232. The method of claim 229, wherein the fixed carbon source is between
83 approximately 4.0% and approximately 9.0% glycerol vol/vol and the cells are cultured in ,84 the absence of light.
!85 233. The method of claim 232, wherein the fixed carbon source is between
186 approximately 5.0% and approximately 8.0% glycerol vol/vol.
!87 234. The method of claim 233, wherein the fixed carbon source is
.88 approximately 7.0% glycerol vol/vol.
£89 235. The method of claim 213, wherein the cells are grown in the presence
£90 of at least 5.0 uM of an exogenously provided fixed carbon source selected from Tables 2
291 and 3.
292 236. A polysaccharide produced from a cell of the genus Porphyridiurn,
293 comprising xylose, glucose, and galactose wherein the molar amount of glucose in the
294 polysaccharide is at least 65% of the molar amount of galactose.
295 237. The polysaccharide of claim 236, wherein the molar amount of
296 glucose in the polysaccharide is at least 75% of the molar amount of galactose.
297 238. The polysaccharide of claim 236, wherein the molar amount of
298 glucose in the polysaccharide is greater than the molar amount of galactose.
299 239. The polysaccharide of claim 236, wherein the polysaccharide is
300 . substantially free of protein.
301 240. A polysaccharide produced from a cell of the genus Porphyridiurn,
302 comprising xylose, glucose, galactose, mannose, and rhamnose, wherein the molar amount
303 of rhamnose in the polysaccharide is at least 2-fold greater than the molar amount of
304 mannose.
305 241. The polysaccharide of claim 240, wherein the polysaccharide is
306 substantially free of protein.
307 242. A polysaccharide produced from a cell of the genus Porphyridiurn,
308 comprising xylose, glucose, galactose, mannose, and rhamnose, wherein the molar amount
309 of mannose in the polysaccharide is at least 2-fold greater than the molar amount of
310 rhamnose.
31 1 243. The polysaccharide of claim 242, wherein the polysaccharide is
312 substantially free of protein.
313 244. A polysaccharide produced from a cell of the genus Porphyridium,
314 comprising xylose, glucose and galactose, wherein the molar amount of galactose in the
315 polysaccharide is greater than the molar amount of xylose.
160
16 245. The polysaccharide of claim 244, wherein the polysaccharide is .
17 substantially free of protein.
18 246. A polysaccharide produced from a cell of the genus Porphyridium,
19 comprising xylose, glucose, glucuronic acid and galactose, wherein the molar amount of ι20 glucuronic acid in the polysaccharide is at least 50% of the molar amount of glucose. 121 247. The polysaccharide of claim 246, wherein the polysaccharide is (22 substantially free of protein.
S 23 248. A polysaccharide produced from a cell of the genus Poφhyridium,
524 comprising xylose, glucose, glucuronic acid, galactose, and at least one monosaccharide
525 selected from the group consisting of arabinose, fucose, N-acetyl galactosamine, and N-
526 acetyl neuraminic acid. 27 249. The polysaccharide of claim 248, wherein the polysaccharide is 28 substantially free of protein. 29 250. A method of producing a nutraceutical composition comprising: 30 a. culturing red microalgae; 31 b. separating the microalgae from culture media; and 32 c. disrupting the microalgae to produce a homogenate. 33 251. The method of claim 250, further comprising drying the microalgae 34 before or after the disrupting step. 35 252. The method of claim 250, further comprising formulating the 36 homogenate with a carrier suitable for human consumption. 37 253. The method of claim 252, wherein the carrier is a plant or animal 38 product. 39 254. The method of claim 252, further comprising formulating the 40 homogenate with a carrier suitable for human oral consumption as a tablet. 41 255. The method of claim 251 , wherein the drying is performed by tray 42 drying, spin drying, rotary drying, spin flash drying, or lyophilization. 43 256. The method of claim 250, wherein the disruption is performed by a 44 method selected from the group consisting of pressure disruption, sonication, jet milling and 45 ball milling. 46 257. The method of claim 250, wherein the red microalgae is of the 47 species Porphyridium.
161
548 258. The method of claim 250, wherein the homogenate contains at least
$49 twice the amount of solvent-available polysaccharide present in a quantity of
550 unhomogenized cells needed to generate the homogenate.
?51 259. The method of claim 280, wherein the homogenate contains at least
352 five times the amount of solvent-available polysaccharide present in a quantity of
353 unhomogenized cells needed to generate the homogenate..
354 260. The method of claim 280, wherein the homogenate contains at least
355 twenty times the amount of solvent-available polysaccharide present in a quantity of
356 unhomogenized cells needed to generate the homogenate.
357 261. The method of claim 250, wherein the microalgae contains an
358 exogenous gene that encodes a protein which either
359 a. increases the production of a small molecule naturally produced
360 by the microalgae; or
361 b. induces the microalgae to produce a small molecule not naturally
362 produced by the microalgae.
363 262. The method of claim 261, wherein the small molecule is a carotenoid.
364 263. The method of claim 262, wherein the carotenoid is lycopene, lutein,
365 beta carotene, or zeaxanthin.
366 264. The method of claim 261 , wherein the small molecule is a
367 polyunsaturated fatty acid.
368 265. The method of claim 264, wherein the polyunsaturated fatty acid is
369 selected from the group consisting of EPA, DHA, linoleic acid and ARA.
370 266. A nutraceutical composition comprising homogenized red microalgae
371 cells and a carrier suitable for human consumption.
372 267. The composition of claim 266, wherein the red microalgae cells
373 contain an exogenous gene that encodes a protein which either:
374 a. increases the production of a small molecule naturally produced
375 by the red microalgae; or
376 b. induces the red microalgae to produce a small molecule not
377 naturally produced by the microalgae.
378 268. The composition of claim 266, containing a small molecule not
379 naturally produced by the microalgae.
380 269. The composition of claim 269, wherein the small molecule is DHA.
162
381 270. The composition of claim 266, further comprising an
382 exopolysaccharide produced by the red microalgae, wherein the exopolysaccharide has been
383 purified from culture media used to grow the red microalgae and is added to the cells
384 before, during, or after homogenization.
385 271. The composition of claim 266, further comprising an exogenously
386 added molecule selected from the list consisting of EPA, DHA, linoleic acid, ARA,
387 lycopene, lutein, beta carotene, and zeaxanthin.
388 272. The composition of claim 266, wherein the homogenized red
389 microalgae cells contain at least twice the amount of solvent-available polysaccharide
390 present in a quantity of unhomogenized cells needed to generate the homogenized red
391 microalgae cells.
392 273. The composition of claim 266, wherein the homogenized red
393 microalgae cells contain at least five times the amount of solvent-available polysaccharide
394 present in a quantity of unhomogenized cells needed to generate the homogenized red
395 microalgae cells.
396 274. The composition of claim 266, wherein the homogenized red
397 microalgae cells contain at least twenty times the amount of solvent-available
398 polysaccharide present in a quantity of unhomogenized cells needed to generate the
399 homogenized red microalgae cells.
400 275. The composition of claim 266, wherein the red microalgae cells are
401 of the genus Porphyridium.
402 276. A composition comprising a purified first polysaccharide produced
403 from a microalgae species listed in Table 1 and a carrier suitable for human consumption.
404 277. The composition of claim 276 wherein the composition contains
405 between 1 microgram and 50 grams of polysaccharide.
406 278. The composition of claim 276, wherein the polysaccharide is
407 sulfated.
408 279. The composition of claim 276, wherein the average molecular weight
409 of the polysaccharide is between 2 and 7 million Daltons.
410 280. The composition of claim 276, wherein the average molecular weight
411 of the polysaccharide is less than 200,000 Daltons.
412 281. The composition of claim 276, further comprising at least one
413 additional polysaccharide.
163
14 282. The composition of claim 281 , wherein the at least one additional
H 5 polysaccharide is from a non-microalgae species.
H 6 283. The composition of claim 282, wherein the at least one additional
U 7 polysaccharide is beta glucan. H 8 284. The composition of claim 276, further comprising a plant phytosterol.
119 285. A composition comprising:
120 a. microalgal cell homogenate; and 1-21 b. a topical carrier. 22 286. The composition of claim 285, wherein the microalgal cell -23 homogenate is produced from a species selected from Table 1. 24 287. The composition of claim 285, wherein the microalgal cell 25 homogenate contains at least twice the amount of solvent-available polysaccharide present 26 in a quantity of unhomogenized cells needed to generate the microalgal cell homogenate. 27 288. The composition of claim 285, wherein the microalgal cell 28 homogenate contains at least five times the amount of solvent-available polysaccharide 29 present in a quantity of unhomogenized cells needed to generate the microalgal cell 30 homogenate. 31 289. The composition of claim 285, wherein the microalgal cell 32 homogenate contains at least twenty times the amount of solvent-available polysaccharide 33 present in a quantity of unhomogenized cells needed to generate the microalgal cell 34 homogenate. 35 290. The composition of claim 285, wherein the red microalgae cells are 36 of the genus Porphyridium . 37 291. A composition comprising purified microalgal polysaccharide and a 38 carrier suitable for topical administration. 39 292. The composition of claim 291 , wherein the purified microalgal 40 polysaccharide is produced from a species selected from Table 1. 41 293. The composition of claim 292, wherein the purified microalgal 42 polysaccharide is produced from a species of the genus Porphyridium. 43 294. The composition of claim 293, wherein the polysaccharide is 44 associated with a fusion protein comprising a first protein with at least 60% amino acid 45 identity with the protein of SEQ ID NO: 28, and a second protein, wherein the fusion 46 protein binds to a sulfated exopolysaccharide from a cell of the genus Porphyridium.
164 .
47 295. The composition of claim. 294, wherein the second protein is selected
1-48 from the group consisting of an antibody and an enzyme.
149 296. The composition of claim 295, wherein the second protein is an
1-50 enzyme.
!-5I 297. The composition of claim 296, wherein the enzyme is a superoxide
1-52 dismutase.
1-53 298. The composition of claim 297, wherein the superoxide dismutase has
154 at least 60% amino acid identity with a member of the group consisting of SEQ ID NO: 14,
Φ55 SEQ ID NO: 15, and a sequence from Table 16, and wherein the composition exhibits
1-56 superoxide dismutase activity.
457 299. The composition of claim 294, wherein the second protein is an
458 antibody.
459 300. The composition of claim 299, wherein the antibody selectively binds 60 to an antigen from a pathogen selected from the group consisting of HIV, Herpes Simplex 61 Virus, gonorrhea, Chlamydia, Human Papillomavirus, and Trichomoniasis. 62 301. The composition of claim 300, wherein the antibody is a humanized 63 antibody.
464 302. The composition of claim 291, wherein the polysaccharide 65 composition contains polysaccharide produced from at least two species selected from 66 Table 1. 67 303. The composition of claim 291, wherein the composition contains 68 between 0.001 % and about 100% polysaccharide by weight. 69 304. A method of formulating a cosmeceutical composition comprising: 70 a. culturing microalgae cells in suspension under conditions to allow
471 cell division; 72 b. separating the microalgae cells from culture media, wherein the 73 culture media contains exopolysaccharide molecules produced by the microalgae cells; 74 c. separating the exopolysaccharide molecules from other molecules
475 present in the culture media;
476 d. homogenizing the microalgae cells; and
477 e. adding the separated exopolysaccharide molecules to the cells
478 before, during, or after homogenization.
479 305. The method of claim 304, wherein microalgae cells are of the genus
480 Porphyridium.
165 1-81 306. A composition containing homogenized microalgae and
^82 exopolysaccharide molecules, wherein the composition is a percentage of
1-83 exopolysaccharide by weight selected from the group consisting of up to 0.1%, up to 1%, up
1-84 to 2%, up to 5%, up to 10%, and up to 20%.
185 307. A combination product comprising:
186 a. a first composition comprising a microalgal homogenate and a ♦87 carrier suitable for topical application; and
188 b. a second composition comprising at least one compound and a
489 carrier suitable for human consumption;
490 wherein the first and second compositions are packaged for sale as a single
491 unit.
492 308. The combination product of claim 307, wherein the first and second
493 compositions contain at least one compound in common.
494 309. The combination product of claim 308, wherein the at least one
495 compound is selected from the group consisting of DHA, EPA, ARA, lycopene, lutein, beta
496 carotene, zeaxanthin, linoleic acid, vitamin C, a polysaccharide, a microalgal homogenate,
497 and superoxide dismutase.
498 310. The combination product of claim 307, wherein the microalgal
499 homogenate is produced from a cell of the genus Porphyridium.
500 311. A method of lowering serum cholesterol in a patient comprising
501 orally administering a polysaccharide produced by microalgae or a microalgal cell
502 homogenate with a biologically acceptable carrier to a patient and thereby lowering the
503 serum cholesterol.
504 312. The method of claim 311, wherein the microalgae is selected from
505 Table 1.
506 313. The method of claim 312, wherein the polysaccharide is produced my
507 microalgae of the genus Porphyridium.
508 314. The method of claim 311, wherein the polysaccharide is
509 administered as component of a food composition.
510 315. The method of claim 311, wherein the amount of polysaccharide
511 administered is from about 0.1 to 50 grams per day.
512 316. The method of claim 315, wherein the amount of polysaccharide
513 administered is from about 0.25 to 6 grams per day.
514 317. A cholesterol lowering composition comprising:
166 i 15 a. a purified microalgal exopolysaccharide or a microalgal cell
>16 homogenate; and il7 b. a carrier suitable for oral consumption.
518 318. The composition of claim 317, wherein the exopolysaccharide or cell
>19 homogenate is produced from cells of the genus Porphyridium.
)20 319. The composition of claim 318, wherein the microalgal cell
521 homogenate contains at least twice the amount of solvent-available polysaccharide present
522 in a quantity of urihomogenized cells needed to generate the microalgal cell homogenate.
523 320. The composition of claim 318, wherein the microalgal cell
524 homogenate contains at least five times the amount of solvent-available polysaccharide
525 present in a quantity of unhomogenized cells needed to generate the microalgal cell
526 homogenate.
527 321. The composition of claim 318, wherein the microalgal cell
528 homogenate contains at least twenty times the amount of solvent-available polysaccharide
529 present in a quantity of unhomogenized cells needed to generate the microalgal cell
530 homogenate.
531 322. The composition of claim 317 comprising an exopolysaccharide,
532 wherein the exopolysaccharide is substantially free of protein.
533 323. The composition of claim 317, wherein the molar amount of glucose
534 in the exopolysaccharide or cell homogenate is at least 55% of the molar amount of
535 galactose in the exopolysaccharide.
536 324. The composition of claim 323, wherein the molar amount of glucose
537 in the exopolysaccharide or cell homogenate is at least 75% of the molar amount of
538 galactose in the exopolysaccharide.
539 325. A sexually transmitted disease prevention kit or composition
540 comprising:
541 a. a solution comprising a polysaccharide produced from
542 microalgae; and
543 b. a prophylactic device.
544 326. The kit or composition of claim 325, wherein the device is a condom
545 that is packaged in direct contact with the solution.
546 327. The kit or composition of claim 325, wherein the composition lacks a
547 lubricant other than polysaccharide.
167
48 328. The kit or composition of claim 325, wherein the polysaccharide
49 provides both lubricant function and antiviral activity.
»50 329. The kit or composition of claim 325, wherein the polysaccharide is
»51 sulfated.
>52 330. The kit or composition of claim 325, wherein the microalgae is
>53 selected from Table 1.
>54 331. The kit or composition of claim 330, wherein the microalgae is
555 selected from the group consisting of Porphyridium sp. and Porphyridium cruentuin.
556 332. The kit or composition of claim 325, wherein the polysaccharide is
557 associated with a fusion protein comprising a first protein with at least 60% amino acid 58 identity with the protein of SEQ ID NO: 28, and a second protein,m and wherein the fusion 59 protein binds a sulfated exopolysacharide from a cell of the genus Porphyridium. 60 333. The kit or composition of claim 332, wherein the second protein is an 61 antibody that selectively binds to an antigen from a pathogen selected from the group 62 consisting of HIV, Herpes Simplex Virus, gonorrhea, Chlamydia, Human Papillomavirus, 63 and Trichomoniasis. 64 334. A method of producing a polysaccharide comprising culturing a 65 microalgae cell in the presence of at least 0.01 micromolar of a compound, wherein the 66 compound is incorporated into the polysaccharide produced by the cell. 67 335. The method of claim 334, wherein the compound is selected from 68 Tables 2 or 3. 69 336. The method of claim 334 wherein the cells are selected from the 70 species listed in Table 1.
571 337. A method of producing an exopolysaccharide comprising
572 a. culturing cells of the genus Porphyridium;
573 b. separating cells from culture media;
574 c. destroying protein attached to the exopolysaccharide present in
575 the culture media; and
576 d. separating the exopolysaccharide from contaminants.
577 338. The method of claim 337, wherein one or more contaminants are
578 selected from the group consisting of amino acids, peptides, proteases, protein fragments,
579 and salts.
168 >80 339. The method of claim 338, wherein at least one contaminant is
581 selected from the group consisting of NaCl, MgSO4, MgCl2, CaCl2, KNO3, KH2PO4,
582 NaHCO3, Tris, ZnCl2, H3BO3, CoCl2, CuCl2, MnCl2, (NH4)6Mo7O24, FeCl3 and EDTA.
583 340. A cell of the genus Porphyridum comprising an exogenous gene that
584 encodes a carbohydrate transporter protein.
585 341. The cell of claim 340, wherein the protein has at least 60% amino
586 acid sequence identity with a protein selected from the group consisting of SEQ ID NOs:
587 20, 22, 24, 26, 27, 29-39 and 46-48.
588 342. The cell of claim 341, wherein the cell contains a nucleic acid that
589 has at least 60% nucleotide identity with a nucleic acid selected from the group consisting
590 of SEQ ID NOs: 21 , 23 and 25.
591 343. A method of trophically converting a cell of the genus Porphyridium
592 comprising:
593 a. providing a nucleic acid encoding a carbohydrate transporter
594 protein;
595 b. transforming the cell with the nucleic acid; and
596 c. selecting for the ability to undergo cell division
597 i. in the absence of light; and
598 ii. in the presence of a carbohydrate that is transported by the
599 carbohydrate transporter protein.
600 344. The method of claim 343, wherein the carbohydrate transporter
601 protein has at least 60% amino acid identity with one or more of SEQ ID NOs: 20, 22, 24,
602 26, 27, 46, 47 and 48.
603 345. The method of claim 343, wherein the nucleic acid encoding a
604 carbohydrate transporter protein is in operable linkage with a promoter active in microalgae.
605 346. The method of claim 343, wherein the carbohydrate is selected from
606 Tables 2 or 3.
607 347. An expression vector comprising a nucleic acid sequence encoding a
608 carbohydrate transporter protein, wherein the nucleic acid has at least 60% nucleotide
609 identity with one or more of SEQ ID NOs: 21, 23 or 25.
610 348. A method of trophically converting a microalgae cell comprising:
611 a. subjecting the cell to a mutagen;
612 b. placing the cell in the presence of a molecule listed in Tables 2 or
613 3; and
169 )14 c. selecting for the ability to undergo cell division in the absence of
515 light.
516 349. The method of claim 348, wherein the cell is of a species listed in
517 Table 1.
518 350. The method of claim 348, wherein the mutagen is selected from the
519 group consisting of ultraviolet light, nitrosoguanidine, and ethane methyl sulfonate.
520 351. The method of claim 348, wherein the cell is of the genus
521 Porphyridium.
622 352. A method of expressing an exogenous gene in a cell of the genus
623 Porphyridium comprising:
624 a. operably linking a gene encoding a protein that confers resistance
625 to the antibiotic zeocin to a promoter active in microalgae to form a resistance cassette;
626 b. operably linking a gene encoding a second protein to a promoter
627 active in microalgae to form a second expression cassette, wherein the resistance cassette
628 and second expression cassette are physically connected to form a dual expression vector;
629 c. transforming the cell with the dual expression vector; and
630 d. selecting for the ability to survive in the presence of at least 2.5
631 ug/ml zeocin.
632 353. The method of claim 352, wherein the second protein is a mammalian
633 growth hormone or a phytase enzyme.
634 354. The method of claim 353, wherein the growth hormone is selected
635 from the group consisting of bovine growth hormone, porcine growth hormone, and equine
636 growth hormone.
637 355. A cell of the genus Porphyridium comprising an exogenous gene
638 encoding a mammalian growth hormone.
639 356. The cell of claim 355, wherein the gene encodes a growth hormone
640 that has at least 60% amino acid identity with one or more of SEQ ID NOs: 17, 18, 19 and
641 Geribank accession numbers or one of Genbank accession numbers AAW67479;
642 AAW67480; AAB64117; BAC07248; AAF61751; AAX77220; ABA55647; ABI97975;
643 BAA06379; AAU93606; AAX31661 ; NP_001003168.
644 357. The cell of claim 355, wherein the mammalian growth hormone is
645 bovine growth hormone.
646 358. The cell of 357, wherein the cell further comprises bovine growth
647 hormone.
170 >48 359. An animal feed comprising cells of the genus Porphyridium, wherein
>49 the cells contain a mammalian growth hormone or a phytase enzyme.
550 360. The animal feed of claim 359, wherein the growth hormone is bovine
551 growth hormone.
552 361. A method of lubricating the joint of a mammal, comprising inj ecting
553 a polysaccharide produced by microalgae into a cavity containing synovial fluid of the
554 mammal.
555 362. The method of claim 361, wherein the polysaccharide is produced by
656 a microalgae listed in Table 1.
657 363. The method of claim 362, wherein the microalgae is of the genus
658 Porphyridium and the polysaccharide is an exopolysaccharide that is substantially free of
659 protein and sterile.
660 364. A method of regulating insulin in a patient comprising administering
661 a polysaccharide produced by microalgae to the patient and thereby regulating the level of
662 insulin in the patient.
663 365. The method of claim 364, wherein the polysaccharide is produced by
664 a microalgae listed in Table 1.
665 366. The method of claim 365, wherein the microalgae is of the genus
666 Porphyridium.
667 367. A method of cosmetic enhancement comprising injecting a
668 polysaccharide produced by microalgae into mammalian skin.
669 368. The method of claim 367, wherein the polysaccharide is produced by
670 a microalgae listed in Table 1.
671 369. The method of claim 368, wherein the microalgae is of the genus
672 Porphyridium and the polysaccharide is an exopolysaccharide that is substantially free of
673 protein and is sterile.
674 370. A method of stabilizing or emulsifying a food composition
675 comprising adding a polysaccharide produced by microalgae into a food composition.
676 371. The method of claim 370, wherein the polysaccharide is produced by
677 a microalgae listed in Table 1.
678 372. The method of claim 371, wherein the microalgae is of the genus
679 Porphyridium.
680 373. A method of treating or effecting prophylaxis of a mammal having or
681 at risk of an undesired inflammatory response comprising administering a polysaccharide
171 582 produced by microalgae to the mammal and thereby treating or effecting prophylaxis of the
683 mammal.
684 374. The method of claim 373, wherein the administration is oral.
685 375. The method of claim 373, wherein the administration is parenteral.
686 376. The method of claim 373, wherein the polysaccharide is produced by
687 a microalgae listed in Table 1.
688 377. The method of claim 373, wherein the microalgae is of the genus
689 Porphyridium.
690 378. A method of producing a glycopolymer comprising
691 a. providing a transgenic cell containing a recombinant gene
692 encoding a monosaccharide transporter; and
693 b. culturing the cell in the presence of at least one monosaccharide,
694 wherein the monosaccharide is transported by the transporter into the cell and is
695 incorporated into the glycopolymer.
696 379. The method of claim 378, wherein the glycopolymer is a
697 polysaccharide.
698 380. The method of claim 378, wherein the cell is a microalgae.
699 381. The method of claim 380, wherein the cell is selected from Table 1.
700 382. The method of claim 381, wherein the cell is of the genus
701 Porphyridium.
702 383. The method of claim 382, wherein the cell is selected from the group
703 consisting of Porphyridium sp. and Porphyridium cruentum.
704 384. The method of claim 378, wherein the polysaccharide is enriched for
705 the at least one monosaccharide compared to an endogenous polysaccharide produced by a
706 non-transgenic cell of the same species.
707 385. The method of claim 384, wherein the monosaccharide is selected
708 from the group consisting of arabinose, fructose, fucose, galactose, glucose, mannose,
709 xylose, glucuronic acid, glucosamine, galactosamine, rhamnose and N-acetyl glucosamine.
710 386. The method of claim 378, wherein the transporter has a lower PCm for
711 glucose than at least one monosaccharide selected from the group consisting of galactose,
712 xylose, glucuronic acid, mannose, and rhamnose.
713 387. The method of claim 378, wherein the transporter has a lower Km for
714 galactose than at least one monosaccharide selected from the group consisting of glucose,
715 xylose, glucuronic acid, mannose, and rhamnose.
172
716 388. The method of claim 378, wherein the transporter has a lower Km
717 for xylose than at least one monosaccharide selected from the group consisting of glucose,
718 galactose, glucuronic acid, mannose, and rhamnose.
719 389. The method of claim 378, wherein the transporter has a lower Km for
720 glucuronic acid than at least one monosaccharide selected from the group consisting of
721 glucose, galactose, xylose, mannose, and rhamnose.
722 390. The method of claim 378, wherein the transporter has a lower Km for
723 mannose than at least one monosaccharide selected from the group consisting of glucose,
724 galactose, xylose, glucuronic acid, and rhamnose.
725 391. The method of claim 378, wherein the transporter has a lower Km for
726 rhamnose than at least one monosaccharide selected from the group consisting of glucose,
727 galactose, xylose, glucuronic acid, and mannose.
728 392. The method of claim 378, wherein the cell is cultured in the presence
729 of at least two monosaccharides, both of which are transporter by the transporter.
730 393. The method of claim 392, wherein the two monosaccharides are
731 selected from the list consisting of glucose, galactose, xylose, glucuronic acid, rhamnose
732 and mannose.
733 394. A method of purifying an exopolysaccharide from cultures of
734 Porphyridiurn comprising:
735 a. culturing Porphyridium cells;
736 b. separating cells from culture media;
737 c. adding isopropanol to the culture media; and
738 d. separating precipitated exopolysaccharide from the solution.
739 395. The method of claim 394, wherein the concentration of isopropanol
740 used to precipitate the exopolysaccharide is between about 20% and 50% vol/vol.
741 396. The method of claim 395, wherein the concentration of isopropanol
742 used to precipitate the exopolysaccharide is between about 30% and 45% vol/vol.
743 397. The method of claim 396, wherein the concentration of isopropanol
744 used to precipitate the exopolysaccharide is between about 36% and 40% vol/vol.
745 398. The method of claim 397, wherein the concentration of isopropanol
746 used to precipitate the exopolysaccharide is about 38.5% vol/vol.
747 399. A system for separating cellular and non-cellular algal biomass
748 production comprising;
749 a. a bioreactor containing microalgae and culture media; and
173 750 b. a first filtration device comprising a filtration unit that maintains
751 cells as retentate and allows exopolysaccharide to pass through as filtrate, wherein the first
752 filtration device is connected to the bioreactor by means for conveying cells and culture
753 media without compromising the axenic nature of the bioreactor; and
754 c. a second filtration device comprising a filtration unit that does not
755 allow exopolysaccharide molecules to pass through;
756 wherein the retentate from the first filtration device and the filtrate from the
757 second filtration device are returned to the bioreactor without compromising the axenic 758 nature of the bioreactor.
759 400. The system of claim 399, wherein the microalgae is of the genus
760 Porphyridium.
761 401. The composition of claim 318, wherein the microalgal cell
762 homogenate contains at least twenty-five times the amount of solvent-available
763 polysaccharide present in a quantity of unhomogenized cells needed to generate the
764 microalgal cell homogenate.
765 . 402. The composition of claim 318, wherein the wherein cell homogenate
766 is produced from cells of the species Porphyridium sp.
767 403. The composition of claim 318, wherein the wherein cell homogenate
768 is produced from cells of the species Porphyridium cruentum.
769 404. A method of formulating a composition comprising
770 a. culturing microalgae cells that produce an exopolysaccharide; and
771 b. concentrating the culture by tangential flow filtration, wherein the
772 exopolysaccharide and cells are retained in the retentate; and
773 c. diafiltering the retentate until the composition is substantially free of
774 salts.
775 405. A method of producing a nutraceutical composition comprising:
776 a. culturing red microalgae;
777 b. separating the microalgae from culture media; and
778 c. disrupting the microalgae to produce a homogenate.
779 406. The method of claim 405, wherein the microalgae contains an
780 exogenous gene that encodes a protein which either
781 a. increases the production of a small molecule naturally produced by the microalgae;
782 or
174 ^83 b. induces the microalgae to produce a small molecule not naturally produced by the f84 microalgae.
785 407. The method of claim 406, wherein the exogenous gene is in operable
786 linkage with a CMV promoter.
787 408. The method of claim 406, wherein the exogenous gene is in operable
788 linkage with a nucleic acid of SEQ ID NO:44 or any fragment thereof.
789 409. The method of claim 406, wherein the exogenous gene is in operable
790 linkage with a nucleic acid of SEQ ID NO:55 or any fragment thereof.
791 410. The method of claim 406, wherein the small molecule is a carotenoid.
792 411. The method of claim 410, wherein the carotenoid is lycopene, lutein,
793 beta carotene, or zeaxanthin.
794 412. The method of claim 406, wherein the small molecule is a
795 polyunsaturated fatty acid.
796 413. The method of claim 412, wherein the polyunsaturated fatty acid is
797 selected from the group consisting of EPA, DHA, linoleic acid and ARA.
798 414. A nutraceutical composition comprising homogenized red microalgae
799 cells and a carrier suitable for human consumption.
800 415. The composition of claim 414, wherein the red microalgae cells
801 contain an exogenous gene that encodes a protein which either:
802 a. increases the production of a small molecule naturally produced by the red
803 microalgae; or
804 b. induces the red microalgae to produce a small molecule not naturally produced by
805 the microalgae.
806 416. The composition of claim 415, wherein the exogenous gene is in
807 operable linkage with a CMV promoter.
808 417. The method of claim 415, wherein the exogenous gene is in operable
809 linkage with a nucleic acid of SEQ ID NO:44 or any fragment thereof.
810 418. The method of claim 415, wherein the exogenous gene is in operable
811 linkage with a nucleic acid of SEQ ID NO:55 or any fragment thereof.
812 419. The composition of claim 414, containing a small molecule not
813 naturally produced by the microalgae.
814 420. The composition of claim 419, wherein the small molecule is DHA.
815 421. The composition of claim 414, further comprising an
816 exopolysaccharide produced by the red microalgae, wherein the exopolysaccharide has been
175 > 17 purified from culture media used to grow the red microalgae and is added to the cells
118 before, during, or after homogenization.
119 422. The composition of claim 414, further comprising an exogenously
320 added molecule selected from the list consisting of EPA, DHA, linoleic acid, ARA,
321 lycopene, lutein, beta carotene, and zeaxanthin.
322 423. The composition of claim 414, wherein the homogenized red
323 microalgae cells contain at least twice the amount of solvent-available polysaccharide
824 present in a quantity of unhomogenized cells needed to generate the homogenized red
825 microalgae cells.
826 424. The composition of claim 414, wherein the homogenized red
827 microalgae cells contain at least five times the amount of solvent-available polysaccharide
828 present in a quantity of unhomogenized cells needed to generate the homogenized red
829 microalgae cells.
830 425. The composition of claim 414, wherein the homogenized red
831 microalgae cells contain at least twenty times the amount of solvent-available
832 polysaccharide present in a quantity of unhomogenized cells needed to generate the
833 homogenized red microalgae cells.
834 426. The composition of claim 414, wherein the red microalgae cells are
835 of the genus Porphyridium.
836 427. A sexually transmitted disease prevention kit or composition
837 comprising:
838 a. a solution comprising a polysaccharide produced from microalgae; and
839 b. a prophylactic device.
840 428. The kit or composition of claim 427, wherein the microalgae is
841 selected from Table 1.
842 429. The kit or composition of claim 428, wherein the microalgae is
843 selected from the group consisting of Porphyridium sp. and Porphyridium cruentum.
844 430. The kit or composition of claim 427, wherein the polysaccharide is
845 associated with a fusion protein comprising a first protein with at least 60% amino acid
846 identity with the protein of SEQ ID NO: 28, and a second protein.
847 431. The kit or composition of claim 430, wherein the fusion protein is
848 encoded by a coding sequence in operable linkage with a CMV promoter.
176 »49 432. The kit or composition of claim 430, wherein the fusion protein is
550 encoded by a coding sequence in operable linkage with a nucleic acid of SEQ ID NO:44 or i
151 any fragment thereof.
$52 433. The kit or composition of claim 430, wherein the fusion protein is
$53 encoded by a coding sequence in operable linkage with a nucleic acid of SEQ ID NO:55 or
354 any fragment thereof.
355 434. The kit or composition of claim 430, wherein the second protein is an
356 antibody that selectively binds to an antigen from a pathogen selected from the group
857 consisting of HIV, Herpes Simplex Virus, gonorrhea, Chlamydia, Human Papillomavirus,
858 and Trichomoniasis.
859 435. A cell of the genus Porphyridurn comprising an exogenous gene that
860 encodes a carbohydrate transporter protein.
861 436. The cell of claim 435, wherein the protein has at least 60% amino
862 acid sequence identity with a protein selected from the group consisting of SEQ ID NOs:
863 20, 22, 24, 26, 27, 29-39, 46, 47 and 48.
864 437. The cell of claim 435, wherein the cell contains a nucleic acid that
865 has at least 60% nucleotide identity with a nucleic acid selected from the group consisting
866 of SEQ ID NOs: 21, 23 and 25.
867 438. The cell of claim 435, wherein the exogenous gene is in operable
868 linkage with a CMV promoter.
869 439. The cell of claim 435, wherein the exogenous gene is in operable
870 linkage with a nucleic acid of SEQ ID NO:44 or any fragment thereof.
871 440. The cell of claim 435, wherein the exogenous gene is in operable
872 linkage with a nucleic acid of SEQ ID NO:55 or any fragment thereof.
873 441. A method of trophically converting a cell of the genus Porphyridium
874 comprising:
875 a. providing a nucleic acid encoding a carbohydrate transporter protein;
876 b. transforming the cell with the nucleic acid; and
877 c. selecting for the ability to undergo cell division in the presence of
878 fixed carbon material as the sole energy source for the cell, wherein the fixed carbon
879 material is a carbohydrate that is transported by the carbohydrate transporter protein.
880 442. The method of claim 441 , wherein the carbohydrate transporter
881 protein has at least 60% amino acid identity with one or more of SEQ ID NOs: 20, 22, 24,
882 26, 27, 46, 47 and 48.
177 >83 443. The method of claim 441 , wherein the nucleic acid encoding a
£84 carbohydrate transporter protein is in operable linkage with a promoter active in microalgae.
585 444. The method of claim 441, wherein the carbohydrate is selected from
$86 Tables 2 or 3.
?87 445. The method of claim 441 , wherein the nucleic acid encoding a
388 carbohydrate transporter protein is in operable linkage with a CMV promoter.
389 446. The method of claim 441, wherein the nucleic acid encoding a
390 carbohydrate transporter protein is in operable linkage with a nucleic acid of SEQ ID NO:44
391 or any fragment thereof.
892 447. The method of claim 441 , wherein the nucleic acid encoding a
893 carbohydrate transporter protein is in operable linkage with a nucleic acid of SEQ ID NO:55
894 or any fragment thereof.
895 448. An expression vector comprising a nucleic acid sequence encoding a
896 carbohydrate transporter protein, wherein the nucleic acid has at least 60% nucleotide
897 identity with one or more of SEQ ID NOs: 21 , 23 or 25.
898 449. The expression vector of claim 448, wherein the nucleic acid
899 sequence encoding a carbohydrate transporter protein is in operable linkage with a CMV
900 promoter.
901 450. The expression vector of claim 448, wherein the nucleic acid
902 sequence encoding a carbohydrate transporter protein is in operable linkage with a nucleic
903 acid of SEQ ID NO:44 or any fragment thereof.
904 451. The expression vector of claim 448, wherein the nucleic acid
905 sequence encoding a carbohydrate transporter protein is in operable linkage with a nucleic
906 acid of SEQ ID NO:55 or any fragment thereof.
907 452. A method of expressing an exogenous gene in a cell of the genus
908 Porphyridium comprising:
909 a. operably linking a gene encoding a protein that confers resistance to the antibiotic
910 zeocin to a promoter active in microalgae to form a resistance cassette;
911 b. operably linking a gene encoding a second protein to a promoter active in
912 microalgae to form a second expression cassette, wherein the resistance cassette and second
913 expression cassette are physically connected to form a dual expression vector;
914 c. transforming the cell with the dual expression vector; and
915 d. selecting for the ability to survive in the presence of at least 2.5 ug/ml zeocin.
178 > 16 453. The method of claim 452, wherein the exogenous gene is in operable
>17 linkage with a nucleic acid of SEQ ID NO:44 or any fragment thereof. ?18 454. The method of claim 452, wherein the exogenous gene is in operable
?19 linkage with a nucleic acid of SEQ ID NO:55 or any fragment thereof.
920 455. The method of claim 452, wherein the second protein is selected from
921 the group consisting of a mammalian growth hormone, a phytase, and a fusion protein
922 containing a first segment that tightly associates with a microalgal polysaccharide and
923 contains a second segment.
924 456. The method of claim 455, wherein the second protein is a mammalian
925 growth hormone selected from the group consisting of bovine growth hormone, porcine
926 growth hormone, and equine growth hormone.
927 457. A cell of the genus Porphyridium comprising an exogenous gene
928 encoding a protein selected from the group consisting of a mammalian growth hormone, a
929 phytase enzyme, an enzyme that catalyzes the formation of a carotenoid or polyunsaturated
930 fatty acid, a carbohydrate transporter, a collagen sύbunit, elastin, and superoxide dismutase.
931 458. The cell of claim 457, wherein the exogenous gene is in operable
932 linkage with a CMV promoter.
933 459. The cell of claim 457, wherein the exogenous gene is in operable
934 linkage with a nucleic acid of SEQ ID NO:44 or any fragment thereof.
935 460. The cell of claim 457, wherein the exogenous gene is in operable
936 linkage with a nucleic acid of SEQ ID NO:55 or any fragment thereof.
937 461. The cell of claim 457, wherein the exogenous gene encodes a
938 mammalian growth hormone that has at least 60% amino acid identity with one or more of
939 SEQ ID NOs: 17, 18 or 19.
940 462. The cell of claim 461, wherein the mammalian growth hormone is
941 bovine growth hormone.
942 ' 463. The cell of 457, wherein the cell further comprises bovine growth
943 hormone.
944 464. An animal feed comprising cells of the genus Porphyridium, wherein
945 the cells contain a mammalian growth hormone or a phytase enzyme.
946 465. The animal feed of claim 464, wherein the growth hormone is bovine
947 growth hormone.
948 466. A method of producing a glycopolymer comprising:
179 •49 a. providing a transgenic cell containing a recombinant gene encoding a
»50 monosaccharide transporter; and
>51 b. culturing the cell in the presence of at least one monosaccharide,
)52 wherein the monosaccharide is transported by the transporter into the cell and is
)53 incorporated into the glycopolymer.
?54 467. The method of claim 466, wherein the recombinant gene is in
?55 operable linkage with a CMV promoter.
?56 468. The method of claim 466, wherein the recombinant gene is in
?57 operable linkage with a nucleic acid of SEQ ID NO:44 or any fragment thereof.
958 469. The method of claim 466, wherein the recombinant gene is in
959 operable linkage with a nucleic acid of SEQ ID NO:55 or any fragment thereof.
960 470. A recombinant nucleic acid comprising at least 25 nucleotides of
961 SEQ ID NO-.44.
962 471. The recombinant nucleic acid of claim 470 comprising at least 28
963 nucleotides of SEQ ID NO:44.
964 472. The recombinant nucleic acid of claim 471 comprising at least 30
965 nucleotides of SEQ ID NO:44.
966 473. The recombinant nucleic acid of claim 472 comprising at least 35
967 nucleotides of SEQ ID NO:44.
968 474. The recombinant nucleic acid of claim 470, .wherein the nucleic acid
969 is operably linked to a heterologous coding sequence.
970 475. The recombinant nucleic acid of claim 474, wherein the heterologous
971 coding sequence encodes a protein selected from the group consisting of and antibody, an
972 antibody fragment, a mammalian growth hormone, a vaccine antigen, a carbohydrate
973 transporter, an antibiotic resistance protein, a fusion protein containing a first segment that
974 tightly associates with a microalgal polysaccharide and contains a second segment, and an
975 enzyme.
976 476. The recombinant nucleic acid of claim 475, wherein the heterologous
977 coding sequence encodes a fusion protein containing a first segment that tightly associates
978 with a microalgal polysaccharide and contains a second segment, wherein the second
979 segment is selected from the group consisting of a collagen chain, elastin, superoxide
980 dismutase, an antibiotic resistance protein, and an antibody.
180 (81 477. The recombinant nucleic acid of claim 474, wherein the heterologous
»82 coding sequence is a carbohydrate transporter selected from the group consisting of SEQ ID
»83 NOs: 20, 22, 24, 26, 27, 46, 47 and 48.
>84 478. The recombinant nucleic acid of claim 474, wherein the heterologous
>85 coding sequence is a hexokinase.
)86 479. A recombinant nucleic acid comprising a first segment that has at
)87 least 25% nucleotide identity with any segment of SEQ ID NO:44, wherein the first
?88 segment is capable of initiating transcription in a Rhodophyta cell.
989 480. The recombinant nucleic acid of claim 479, wherein the first segment
990 has a percent nucleotide identity with any segment of SEQ ID NO:44 selected from the
991 group consisting of 35%, 45%, 55%, 65%, 75%, 85%, 95% and 98%.
992 481. The recombinant nucleic acid of claim 479, wherein the first segment
993 is capable of initiating transcription in a Bangiophyceae cell.
994 482. The recombinant nucleic acid of claim 481, wherein the first segment
995 is capable of initiating transcription in a Porphyridiales cell.
996 483. The recombinant nucleic acid of claim 482, wherein the first segment
997 is capable of initiating transcription in a Porphyridiaceae cell.
998 484. The recombinant nucleic acid of claim 484, wherein the first segment
999 is capable of initiating transcription in a Porphyridium cell.
000 485. A recombinant nucleic acid comprising a non-transcribed promoter
001 region of a gene in the nuclear genome of a cell of the genus Porphyridium.
L 002 486. The recombinant nucleic acid of claim 485, wherein the promoter
1003 region is SEQ ID NO:44 or a fragment thereof.
[004 487. A cell of the genus Porphyridium containing a recombinant copy of
1005 any segment of SEQ ID NO:44.
1006
181
EP07808975A 2006-01-19 2007-01-19 Nutraceutical compositions from microalgae and related methods of production and administration Withdrawn EP1993565A2 (en)

Applications Claiming Priority (12)

Application Number Priority Date Filing Date Title
US11/336,426 US20070191303A1 (en) 2006-01-19 2006-01-19 Polysaccharide compositions and methods of producing, screening, and formulating polysaccharide compositions
US11/337,103 US20070166266A1 (en) 2006-01-19 2006-01-19 Methods and compositions for improving the health and appearance of skin
US11/336,431 US20070166449A1 (en) 2006-01-19 2006-01-19 Methods and compositions for thickening, stabilizing and emulsifying foods
US11/337,171 US20070167398A1 (en) 2006-01-19 2006-01-19 Methods and compositions for reducing inflammation and preventing oxidative damage
US11/336,430 US20070167397A1 (en) 2006-01-19 2006-01-19 Methods and compositions for joint lubrication
US11/336,428 US20070167396A1 (en) 2006-01-19 2006-01-19 Methods and compositions for cholesterol reduction in mammals
US11/336,656 US20070166797A1 (en) 2006-01-19 2006-01-19 Devices and solutions for prevention of sexually transmitted diseases
US81696706P 2006-06-28 2006-06-28
US83209106P 2006-07-20 2006-07-20
US83845206P 2006-08-17 2006-08-17
US87207206P 2006-11-30 2006-11-30
PCT/US2007/001319 WO2007136428A2 (en) 2006-01-19 2007-01-19 Nutraceutical compositions from microalgae and related methods of production and administration

Publications (1)

Publication Number Publication Date
EP1993565A2 true EP1993565A2 (en) 2008-11-26

Family

ID=38288316

Family Applications (3)

Application Number Title Priority Date Filing Date
EP18165955.8A Active EP3398606B1 (en) 2006-01-19 2007-01-19 Microalgae-derived compositions for improving the health and appearance of skin
EP07718342.4A Active EP1986665B1 (en) 2006-01-19 2007-01-19 Microalgae-derived compositions for improving the health and appearance of skin
EP07808975A Withdrawn EP1993565A2 (en) 2006-01-19 2007-01-19 Nutraceutical compositions from microalgae and related methods of production and administration

Family Applications Before (2)

Application Number Title Priority Date Filing Date
EP18165955.8A Active EP3398606B1 (en) 2006-01-19 2007-01-19 Microalgae-derived compositions for improving the health and appearance of skin
EP07718342.4A Active EP1986665B1 (en) 2006-01-19 2007-01-19 Microalgae-derived compositions for improving the health and appearance of skin

Country Status (5)

Country Link
EP (3) EP3398606B1 (en)
ES (1) ES2682455T3 (en)
IL (1) IL192911A (en)
PL (1) PL3398606T3 (en)
WO (2) WO2007136428A2 (en)

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8277849B2 (en) 2006-01-19 2012-10-02 Solazyme, Inc. Microalgae-derived compositions for improving the health and appearance of skin
US8298548B2 (en) 2007-07-18 2012-10-30 Solazyme, Inc. Compositions for improving the health and appearance of skin
EP2164865A2 (en) * 2007-06-13 2010-03-24 Hochschule Mannheim Compounds for the modulation of huntingtin aggregation, methods and means for identifying such compounds
US8927522B2 (en) 2008-10-14 2015-01-06 Solazyme, Inc. Microalgal polysaccharide compositions
WO2010054322A1 (en) 2008-11-07 2010-05-14 Solazyme, Inc. Cosmetic compositions comprising microalgal components
AU2010295488B2 (en) * 2009-09-18 2016-07-14 Phycoil Biotechnology International, Inc. Microalgae fermentation using controlled illumination
US10479969B2 (en) 2010-10-11 2019-11-19 Phycoil Biotechnology International. Inc. Utilization of wastewater for microalgal cultivation
EP2658399B2 (en) 2010-12-31 2024-02-28 Abbott Laboratories Nutritional compositions comprising human milk oligosaccharides for use in treating and/or preventing enteric rotavirus infection
SG10202110501RA (en) 2010-12-31 2021-11-29 Abbott Lab Methods For Decreasing The Incidence Of Necrotizing Enterocolitis In Infants, Toddlers, Or Children Using Human Milk Oligosaccharides
NZ612105A (en) 2010-12-31 2015-02-27 Abbott Lab Human milk oligosaccharides for modulating inflammation
SG191394A1 (en) 2010-12-31 2013-08-30 Abbott Lab Methods for reducing the incidence of oxidative stress using human milk oligosaccharides, vitamin c and anti-inflammatory agents
CN103391783A (en) 2010-12-31 2013-11-13 雅培制药有限公司 Methods of using human milk oligosaccharides for improving airway respiratory health
CA2822500C (en) 2010-12-31 2022-07-26 Abbott Laboratories Human milk oligosaccharides to promote growth of beneficial bacteria
WO2012092157A2 (en) 2010-12-31 2012-07-05 Abbott Laboratories Nutritional formulations including human milk oligosaccharides and antioxidants and uses thereof
PT2495306T (en) * 2011-03-04 2017-10-05 Butalco Gmbh Specific arabinose transporter of the plant arabidosis thaliana for the construction of pentose-fermenting yeasts
WO2013003268A1 (en) 2011-06-27 2013-01-03 Dow Global Technologies Llc Genetically-engineered microbial oil dielectric fluid
WO2013032674A1 (en) 2011-08-29 2013-03-07 Abbott Laboratories Human milk oligosaccharides for preventing injury and/or promoting healing of the gastrointestinal tract
AU2013251382A1 (en) 2012-04-27 2014-10-16 Synthetic Genomics, Inc. Compositions and methods for modulating the sensitivity of cells to AHAS inhibitors
US9125911B2 (en) * 2013-03-14 2015-09-08 Quadex Pharmaceuticals, Llc Combined systemic and topical treatment of disordered tissues
WO2014186395A1 (en) 2013-05-15 2014-11-20 Solazyme, Inc. Cosmetic compositions comprising microalgal oil
MD4334C1 (en) * 2014-02-03 2015-10-31 Государственный Университет Молд0 Strain of blue-green microalga Cylindrospermum licheniforme (Bory) Kütz - source of glucides
MD4351C1 (en) * 2014-04-08 2016-01-31 Государственный Университет Молд0 Strain of blue-green microalga Calothrix elenkinii Kossinsk. - source of glucides
CN104072543A (en) * 2014-04-22 2014-10-01 浙江海洋学院 Galactofuranose as well as preparation method and application thereof
EP3288388A1 (en) * 2015-04-30 2018-03-07 Micoperi Blue Growth S.r.l. Microalgae extract for agricultural use
EP3340976A4 (en) * 2015-08-28 2019-04-17 Algahealth (AH) Ltd. Improved process for producing fucoxanthin and/or polysaccharides from microalgae
US10675233B2 (en) * 2015-11-18 2020-06-09 Seiberg Consulting, LLC Compositions containing natural extracts and use thereof for skin and hair
CN106977439B (en) * 2017-03-23 2018-10-02 广州智特奇生物科技股份有限公司 A kind of isomerization method of lutein
CN107043800A (en) * 2017-05-13 2017-08-15 广州固仑生物科技有限公司 A kind of preparation method of collagen
US11020340B2 (en) 2017-09-18 2021-06-01 Seiberg Consulting, LLC Compositions containing natural products and use thereof for skin and hair
CN109400745B (en) * 2018-11-21 2021-01-05 温州大学苍南研究院 Preparation of low molecular weight porphyra yezoensis polysaccharide and application of low molecular weight porphyra yezoensis polysaccharide in resisting human cervical cancer cell tumor
CN113278091A (en) * 2021-06-29 2021-08-20 广东湛江海洋医药研究院 Porphyridium polysaccharide and preparation method and application thereof
CN115671368B (en) * 2022-10-28 2024-03-22 东华大学 Nanofiber composite patch for removing biological membrane and preparation method thereof

Family Cites Families (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4417415A (en) * 1982-04-26 1983-11-29 Battelle Development Corporation Process for culturing a microalga, and extracting a polysaccharide therefrom
US4992540A (en) 1984-11-28 1991-02-12 Massachusetts Institute Of Technology Glucan composition and process for preparation thereof
US4810646A (en) 1984-11-28 1989-03-07 Massachusetts Institute Of Technology Glucan compositions and process for preparation thereof
US4752496A (en) 1986-05-27 1988-06-21 Qmax Technology Group, Inc. Method of applying cosmetics to a substrate and article
CA1332650C (en) * 1987-06-18 1994-10-18 Kunitaka Hirose Polysaccharides and antiviral drugs containing the same as active ingredient
FR2621327B1 (en) * 1987-10-06 1990-01-05 Commissariat Energie Atomique PROCESS FOR PRODUCING AND EXTRACTING POLYSACCHARIDES FROM A CULTURE OF PORPHYRIDIUM CRUENTUM AND DEVICE FOR CARRYING OUT THIS PROCESS
FR2686619B1 (en) * 1992-01-28 1995-07-13 Commissariat Energie Atomique PROCESS FOR THE SELECTIVE PRODUCTION OF POLYUNSATURATED LIPIDS FROM A CULTURE OF PORPHYRIDIUM MICROALGAE AND TANK USED IN THIS PROCESS.
IL102189A (en) 1992-06-12 1995-07-31 Univ Ben Gurion Microorganism growth apparatus
IL110396A (en) 1994-07-21 2000-07-16 Univ Ben Gurion Coloring materials derived from red microalgae their preparation and compositions containing them
AU697323B2 (en) * 1995-03-23 1998-10-01 Lancaster Group Gmbh Cosmetic with condensates of plant and animal decomposition products
IL114267A0 (en) * 1995-06-22 1995-10-31 Univ Ben Gurion Antiviral agents
WO1997039106A1 (en) 1996-04-12 1997-10-23 Martek Biosciences Corporation Methods and tools for transformation of eukaryotic algae
US6855365B2 (en) 1997-08-13 2005-02-15 Diversa Corporation Recombinant bacterial phytases and uses thereof
US5876997A (en) 1997-08-13 1999-03-02 Diversa Corporation Phytase
US20010055627A1 (en) * 1997-09-26 2001-12-27 Najla Guthrie Compositions And Methods For Regulating Lipoproteins And Hypercholesterolemia With Limonoids, Flavonoids And Tocotrienols
WO2000075282A1 (en) * 1999-06-08 2000-12-14 Aquamer (Saem) Method for obtaining from a culture medium of microalgae, a heat-stable extract with antioxidant and wound healing activity
JP3271008B2 (en) 1999-11-18 2002-04-02 独立行政法人産業技術総合研究所 Method for producing chondroitin sulfates
AU5369901A (en) * 2000-04-21 2001-11-07 Martek Biosciences Corp Trophic conversion of obligate phototrophic algae through metabolic engineering
US6969705B2 (en) 2000-07-21 2005-11-29 Aventis Pharma S.A. Compositions of polysaccharides derived from heparin, their preparation and pharmaceutical compositions containing them
JP2002069443A (en) * 2000-08-30 2002-03-08 Microalgae Corporation Antioxidant and cosmetic containing the antioxidant
US20030207947A1 (en) * 2001-03-07 2003-11-06 Desouza Mervyn L. Production of lutein in microorganisms
EP1264893A1 (en) * 2001-06-08 2002-12-11 Teagasc Dairy Products Research Centre CLA biosynthesis by bifidobacteria
US6680062B2 (en) * 2001-10-05 2004-01-20 Color Access, Inc. Anti-irritating rosacea treatment
DE10151155A1 (en) * 2001-10-19 2003-05-08 Nutrinova Gmbh Native PUFA triglyceride mixtures with a high content of polyunsaturated fatty acids as well as processes for their production and their use
AU2002352337A1 (en) * 2001-11-14 2003-05-26 Larena Product containing a red alga extract of the genus porphyra and uses thereof for protecting cells
US7144595B2 (en) * 2001-11-16 2006-12-05 Brandeis University Prepared foods containing triglyceride-recrystallized non-esterified phytosterols
ATE392472T1 (en) * 2002-02-28 2008-05-15 Takara Bio Inc PROMOTOR AND INTRON FROM ALGAE
US7135290B2 (en) 2003-04-12 2006-11-14 Solazyme, Inc. Methods and compositions for evolving hydrogenase genes
US6986323B2 (en) * 2002-11-25 2006-01-17 Algal Technologies, Inc. Inland aquaculture of marine life using water from a saline aquifer
WO2006013572A2 (en) 2004-08-05 2006-02-09 Ben Gurion University Of The Negev Research And Development Authority Red microalgae expressing exogenous polypeptides and methods of generating and utilizing same
IL172477A (en) * 2005-12-08 2012-06-28 Univ Ben Gurion Viscosupplementation with algal polysaccharides in the treatment of arthritis

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2007136428A2 *

Also Published As

Publication number Publication date
EP1986665A2 (en) 2008-11-05
ES2682455T3 (en) 2018-09-20
PL3398606T3 (en) 2022-02-07
EP1986665A4 (en) 2012-12-05
EP1986665B1 (en) 2018-05-02
IL192911A (en) 2016-05-31
WO2007084769A3 (en) 2008-11-06
WO2007136428A8 (en) 2009-07-23
WO2007136428A3 (en) 2008-11-27
WO2007084769A2 (en) 2007-07-26
EP3398606B1 (en) 2021-09-15
IL192911A0 (en) 2009-02-11
WO2007136428A2 (en) 2007-11-29
EP3398606A1 (en) 2018-11-07

Similar Documents

Publication Publication Date Title
US20090274736A1 (en) Nutraceutical Compositions From Microalgae And Related Methods of Production And Administration
WO2007136428A2 (en) Nutraceutical compositions from microalgae and related methods of production and administration
US10493007B2 (en) Microalgae-derived compositions for improving the health and appearance of skin
US20070191303A1 (en) Polysaccharide compositions and methods of producing, screening, and formulating polysaccharide compositions
US20070167396A1 (en) Methods and compositions for cholesterol reduction in mammals
US20070166266A1 (en) Methods and compositions for improving the health and appearance of skin
US10231907B2 (en) Compositions for improving the health and appearance of skin
US20070167398A1 (en) Methods and compositions for reducing inflammation and preventing oxidative damage
US11744847B2 (en) Use of beta-1,3-glucan for modulating immune function and treating intestinal inflammation
US20070167397A1 (en) Methods and compositions for joint lubrication
US10278912B2 (en) Microalgal polysaccharide compositions
US20070166449A1 (en) Methods and compositions for thickening, stabilizing and emulsifying foods
ES2718275T3 (en) Microalgae polysaccharide compositions
US20070166797A1 (en) Devices and solutions for prevention of sexually transmitted diseases
KR100727339B1 (en) Remedies
BR112018008401B1 (en) USE OF A COMPOSITION
KR20070018147A (en) Remedies

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK RS

R17D Deferred search report published (corrected)

Effective date: 20081127

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 36/00 20060101AFI20090127BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20080820