WO2009080794A1 - Procédé de préparation de matrices extracellulaires spécifiques de cellules - Google Patents

Procédé de préparation de matrices extracellulaires spécifiques de cellules Download PDF

Info

Publication number
WO2009080794A1
WO2009080794A1 PCT/EP2008/068110 EP2008068110W WO2009080794A1 WO 2009080794 A1 WO2009080794 A1 WO 2009080794A1 EP 2008068110 W EP2008068110 W EP 2008068110W WO 2009080794 A1 WO2009080794 A1 WO 2009080794A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
human
stem cells
extracellular matrix
tissue
Prior art date
Application number
PCT/EP2008/068110
Other languages
English (en)
Inventor
Marwan El-Sabban
Steen Sindet-Pedersen
Original Assignee
Mc2 Cell Aps
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
Application filed by Mc2 Cell Aps filed Critical Mc2 Cell Aps
Publication of WO2009080794A1 publication Critical patent/WO2009080794A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/067Hepatocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0663Bone marrow mesenchymal stem cells (BM-MSC)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/90Substrates of biological origin, e.g. extracellular matrix, decellularised tissue

Definitions

  • the invention is in the field of stem cell technology.
  • This invention is directed to an in vitro method for inducing cells of human origin to produce cell-specific extracellular matrices and differentiating factors.
  • This extract is suitable for use in differentiating human stem cells into desired tissue cells, such as differentiating bone marrow-derived stem cells into chondrocytes.
  • Some cell-scaffold compositions have multiple layers of biocompatible materials including extracellular matrix materials such as collagen, fibril-forming collagen, Matrix GIa protein, osteocalcin, or other biocompatible materials including marine coral, coralline hydroxyapatite, ceramic, and mixtures thereof, and some such scaffolds have been seeded with cells, and then placed within a bioreactor having a means for mechanically stimulating the cells at distinct frequencies (see U.S. Patent Application No. 20040005297 to P. R. Connelly et al, filed July 8, 2002, published January 8, 2004).
  • LTEs living tissue equivalents
  • connective tissue e.g., "Apligraf" from Organogenesis
  • LTEs have several advantages over synthetic alternatives including being a natural cell substrate, allowing cellularity to be achieved directly, and being conducive to cell spreading and extracellular matrix (ECM) formation.
  • LTEs are made by mixing cells with a soluble biopolymer solution (e.g., collagen, fibrin, and/or proteoglycans). The cells invade, rearrange and partially degrade the biopolymer scaffold as well as synthesize new proteins throughout the culture period.
  • a soluble biopolymer solution e.g., collagen, fibrin, and/or proteoglycans
  • the cells invade, rearrange and partially degrade the biopolymer scaffold as well as synthesize new proteins throughout the culture period.
  • LTEs generally lack the physical properties necessary to resist in vivo mechanical forces, and are not true "living tissues”.
  • LTEs that are completely cell-derived have been developed. However, to date they have been very thin and taken a long time to grow, generally on the order of months, whereas collagen gels and fibrin gels can be developed in only a few days. There is a pressing need for complete biological cell-derived LTEs, and living scaffolds for use in wound repair and tissue regeneration in vitro and in vivo.
  • ECM extracellular matrix
  • An important component of the cellular environment is the extracellular matrix (ECM), which is composed of glycoproteins, proteoglycans and glycosaminoglycans that are secreted and assembled locally into an organized network to which cells adhere.
  • ECM is a component of the environment of all cell types, although the composition of the ECM and the spatial relationships between cells and ECM differ between tissues.
  • Cells may be completely surrounded by ECM, as is the case for chondrocytes, or may contact the ECM only at one surface, as exemplified by epithelial and endothelial cells. In some tissues only a proportion of the cells are exposed to ECM: for example, in stratified epithelia.
  • ECM extracellular matrix
  • ECM ECM
  • cell growth and differentiation ECM
  • soluble factors that control cell fate.
  • stem cells may therefore ultimately be used as a renewable source of cells that differentiate into a variety of tissue cells useful for treating a number of diseases and deficiencies.
  • One important use is the treatment of neurological diseases such as Parkinson's disease ("PD").
  • PD Parkinson's disease
  • neural stem cells are not a particularly abundant source because they reside deep in the brain, severely constraining accessibility for harvesting.
  • BM bone marrow
  • BM comprises a number of stem cell types. Best known among these are hematopoietic stem cells (HSCs) and marrow stromal cells (MSCs). In normal mammals, HSCs give rise to blood cells whereas MSCs give rise to cell types that populate other tissues and sites such as cartilage or bone, hematopoietic supportive stromal cells and fat. Recent studies have suggested that these BM stem cells can, under appropriate conditions, differentiate into additional cell types such as cardiac myocytes, liver cells, and skeletal muscle cells.
  • HSCs hematopoietic stem cells
  • MSCs marrow stromal cells
  • BM stem cells have been shown to have the potential for generating neurons (Sanchez-Ramos et al. Exp. Neurol. 164 247-256 (2000), Woodbury et al. J: Neurosci. Res. 62: 364-370 (2000), Mezey et al. Science 290: 1779-1782 (2000), Brazelton et al. Science 290: 775-1779 (2000).
  • Chopp's group has investigated the use of human MSCs (hMSCs) to treat rats subjected to strokes. Li Y et al., Neurology, 2002, 59: 514-523, tested the effect of intravenously administered hMSCs on neurologic functional deficits after stroke.
  • BDNF brain- derived neurotrophic factor
  • NGF nerve growth factor
  • VEGF nerve growth factor
  • bFGF basic fibroblast growth factor
  • HGF hepatocyte growth factor
  • hMSCs in such cultures responded by producing more BDNF, NGF, VEGF, and HGF, supporting the notion that transplanted hMSCs provide therapeutic benefit in part via a responsive secretion of an array of growth factors that can foster neuro- protection and angiogenesis.
  • Laboratory grown cells derived from a several stem cell types may be a desirable source of transplantable material for grafting into brains of individuals suffering from neurological disorders.
  • stem cells To induce stem cells to differentiate, it is desirable to identify the right combination of molecules, their relative abundance and cell-culture conditions to (a) support survival and/or self-renewal of undifferentiated cells in culture and (b) stimulate them to become committed to a desired cell lineage. Such cells may then be implanted into an appropriate site in vivo to complete their growth and differentiation program.
  • HSC extracellular matrix
  • ECM extracellular matrix
  • the process of HSC (or other stem cell) differentiation into particular progeny in vitro requires the action of many factors, including growth factors, extracellular matrix (“ECM”) molecules and components, environmental stressors and direct cell-to-cell interactions.
  • ECM extracellular matrix
  • hLIF human "leukemia inhibitory factor”
  • the present invention is directed at the production of extracellular matrix components and soluble factors based on cultivating differentiating stem cells in order to induce their production of extracellular matrix.
  • the present inventors have surprisingly found that human extracellular matrix extracts produced by differentiating stem cells largely reflect the corresponding in vivo composition of the extracellular matrix, and can thus later function as optimized differentiation environment for progenitors and stem cells, which will then differentiate into the cell types that are normally harboring the tissue having this extra cellular matrix composition.
  • the present invention provides a method for manufacturing extracellular extracts and soluble factors ex vivo, wherein the composition of the extracts resembles the human in vivo composition.
  • Such extracts are very suitable for differentiating stem cells into the cells of a tissue of interest.
  • the extracellular matrix resembling the extracellular matrix of the desired tissue may be of human or animal origin, such as equine, canine, porcine, bovine, and ovine sources; or rodent species, such as mouse or rat.
  • an animal extracellular matrix extract may be used to induce the differentiation and thus stimulate the human stem cells to secrete an extracellular matrix of human origin.
  • the human stem cells differentiate they will produce an extracellular matrix layer or body, which predominantly contains differentiated human cells along with the extracellular matrix produced by them-selves; thus, if the stem cells have been stimulated to differentiate in an extract of animal origin there will be no fractions of non-human material left in the produced extracellular matrix layer. Since the in vivo composition of the extracellular matrix is achieved when the stem cells are fully differentiated into the cells of the tissue of interest it is necessary to cultivate the stem cells for generally at least 14 days.
  • the cells are assessed before harvesting the extracellular matrix; as a general rule at least 90% of the cells should be fully differentiated before preparing the extracellular matrix.
  • the stem cells are only initially stimulated with the animal derived extracellular matrix, thereafter the stimulated cells are carefully washed before they are cultured in a common growth medium; ii) the stem cells are stimulated with an extracellular matrix of human origin (difficult to obtain due to pain and regulatory hurdles), and iii) the stem cells are stimulated with a "synthetic" extracellular matrix comprising growth and differentiation factors known to be (at least partially) responsible for the differentiation of stem cells into a given tissue cell type.
  • the present invention is further directed to the use of extracts according to the present invention for differentiation of stem cells.
  • the extracts may be used for differentiation of mesenchymal stem cells into at least one type of tissue, in particular into bone or cartilage tissue.
  • the extracts may be used for differentiation of hematopoietic stem cells into hematopoietic progenitor cells.
  • the present method is used to manufacture extracellular matrix pertaining to cartilage tissue by seeding and culturing stem cells in a medium resembling the composition of the extracellular matrix of cartilage tissue for stimulated synthesis of cartilage extracellular matrix extract.
  • This ECM extract can be directly applied to adult stem cells to differentiate them into chondrocytes.
  • the present invention also provides a method for differentiating human bone marrow-derived stem cells into chondrocytes, comprising:
  • the present method is used to manufacture extracellular matrix pertaining to liver tissue by seeding and culturing hepatocytes for stimulated synthesis of extracellular matrix extract resembling the composition of the matrix in liver tissue.
  • This ECM extract can be directly applied to adult stem cells to differentiate them into hepatocytes.
  • the present invention also provides a method for differentiating bone human marrow-derived stem cells into hepatocytes, comprising:
  • the present method is used to manufacture extracellular matrix pertaining to pancreas tissue, particularly islets tissue, by seeding and culturing pancreas beta-cells for stimulated synthesis of extracellular matrix extract resembling the composition of the matrix in islet tissue.
  • This ECM extract can be directly applied to adult stem cells to differentiate them into beta cells.
  • the present invention also provides a method for differentiating bone human marrow-derived stem cells into beta-cells, comprising:
  • stem cell therapy offers an effective and less invasive alternative to treat bone fractures, or enhance the repair of tissues that require prolonged time to fully heal.
  • Bone marrow-derived mesenchymal stem cells are a population of adult multipotent stem cells located within the bone marrow. These cells are characterized by their ability to differentiate into at least three phenotypes (chondrocytes, osteocytes and adipocytes) when cultured in vitro (Jaiswal 2000 and Pittenger 1999). Once uncommitted mesenchymal stem cells are isolated and purified from the bone marrow they maintain the capacity to self-renew and differentiate along multiple pathways resulting in the generation and maintenance of a variety of tissues. This has advanced the possibility of utilizing these cells to repair or replace damaged tissues.
  • ECM extracellular matrix
  • Figure 1 shows negative cytotoxic effect of cartilage matrix extracts on cultured BMMSCs (A). Decrease in cellular proliferation as an indication of onset of differentiation: BMMSC were treated with cartilage matrix extracts. Cellular proliferation was monitored at day 1 , day 3, day 7 and day 14 post treatment. Upon treatment with organ-derived extracts there was a significant decrease in cellular proliferation. Untreated BMMSC maintained a normal proliferation rate (B).
  • Figure 2 shows the expression of Sox9 using real time PCR technique: Joint and meniscus extracts induce chondrogenic differentiation of BMMSCs through the upregulation of Sox9 mRNA (A).
  • Figure 3 shows the expression of Collagen type I using real time PCR technique. Collagen type I, which is known to be upregulated during osteogenic differentiation, was not detected (B).
  • Figure 4 shows aggrecan expression: Real-time PCR revealed decreased levels of aggrecan mRNA (suggesting late onset of aggrecan expression) (A).
  • Figure 5 relates to aggrecan positive cells, which showed major increase in aggrecan positive cells (C).
  • Figure 6 shows morphological changes during BMMSC chondrocytic differentiation 14 days post treatment: showing aggregates (arrows) with central alignment of hypertrophic cells. 10ng/ml TGF- ⁇ 1 recombinant protein was used as positive control.
  • Figure 7 shows bovine and equine bone extracts (Colloss ® and Colloss-E ® ) induce in vitro differentiation of BMMSCs.
  • BMMSCs at passage six were treated with a 1 :50 dilution of Colloss (bovine) or Colloss-E (equine) (28 and 32 ⁇ g/ml, respectively) for different time points.
  • FIG 8 shows that Colloss ® and Colloss-E ® induce alkaline phosphatase activity in BMMSCs.
  • BMMSCs at passage six were treated with a 1 :50 dilution of Colloss (bovine) or Colloss-E (equine) (28 and 32 ⁇ g/ml, respectively) for 7 or 14 days.
  • (B) Alkaline phosphatase enzyme activity was measured in medium-free cell extracts in the absence or presence of Colloss or Colloss-E at day 7 and day 14. Results are expressed as U/ml/h/ ⁇ g protein (n 2).
  • FIG. 9 shows that Colloss ® and Colloss-E ® induce osteopontin expression in BMMSCs.
  • BMMSCs at passage six were treated with a 1 :50 dilution of Colloss (bovine) or Colloss-E (equine) (28 and 32 ⁇ g/ml, respectively) for 7, 14 and 30 days.
  • BMMSC Bone marrow-derived mesenchymal stem cell.
  • Figure 10 shows that Colloss ® and Colloss-E ® induce calcium deposition in BMMSCs.
  • Figure 1 1 shows typical hepatocyte markers induced in BMMSC
  • Figure 12 shows the induction of p450 expression in ECM differentiated BMMSCs.
  • Figure 13 shows the morphology of differentiated BMMSCs cultivated on human liver cell ECM.
  • FIG 14 shows expression of Liver-specific connexins in differentiated stem cells (hepatocytes).
  • Figure 15 shows Induction of p450 in liver-differentiated BMMSC.
  • Figure 16 shows glucose uptake in liver-differentiated BMMSC compared to undifferentiated BMMSC.
  • the culture media or extracts of the present invention exhibit many of the native features of the tissue from which their cells are derived.
  • “Resembles” as used herein means there is physical, compositional, structural, functional, phenotypic or other similarities between the materials or systems being compared, such that the objects are substantially equivalent. "Substantially equivalent” means that visible, microscopic, physical, functional, and other observations and assays do not easily or significantly distinguish the materials or systems. An easy or significant distinction would, for example, be a functional difference, a physical difference, a compositional difference, a structural difference immediately apparent, or easily detectable with standard assays and observational techniques such as staining, microscopy, antibodies, etc.
  • ECM Extracellular Matrix
  • CDM Cell Derived Matrix
  • Cell-produced Matrix as used interchangeably herein means a cell-derived secreted substance produced by and/or secreted from cells into the extracellular space.
  • the ECM/CDM provides a growth template for any cell type to grow, differentiate, and produce tissue.
  • the ECM allows cell attachment and cell migration, and promotes cell differentiation.
  • the ECM also aids the formation of new tissue of a desired or existing cell type. As used herein
  • Cell-Produced Matrix also called Cell-Derived Matrix (CDM)
  • CDM Cell-Derived Matrix
  • Construct as used herein means a physical structure with mechanical properties such as a matrix of scaffold. Construct encompasses both autogenic living scaffolds and living tissue matrices, ex-vivo cell- produced tissue and cell-derived matrix.
  • Cell-derived as used herein means that the source for the material, body, or component is a cell or a collection of cells.
  • Ex-vivo Cell-produced Tissue means, a functional tissue comprising one or more types of cells (or entities) and the ECM (or matrix) that has been completely produced and arranged by some of these cells (or entities).
  • LTM Local Tissue Matrix
  • LTM means, a 3-dimensional tissue (or matrix) that is capable of being transformed into a more complex tissue (or matrix) or a completely different type of tissue (or matrix) that consists of cells (or entities) and the ECM (or matrix) that has been completely produced and arranged by these cells (or entities).
  • LTE Local Tissue Equivalent
  • “Culturing the cells in order to differentiate them” means conditions that facilitate, aid, further or in any way allow the development of three-dimensional tissue growth. Conditions may include use of specific media, growth factors, minerals, incubation temperature, cell density, aeration, agitation, use of ALS "molds" to shape and contain growth of desired tissue, use of sub-atmospheric pressure chambers such as Synthecon's near-zero-gravity incubator systems (such as HARVs and STLVs) for growth of desired tissue, use of micro-carrier beads, use of natural or biodegradable scaffolds, implanting a non-fibroblast-seeded autogenic living scaffold within an in vivo site such as in an organ or tissue such as connective, epithelial, muscle, and/or nerve tissue.
  • Conditions may include use of specific media, growth factors, minerals, incubation temperature, cell density, aeration, agitation, use of ALS "molds" to shape and contain growth of desired tissue, use of sub-atmosphe
  • Genetically engineered means that a cell or entity, by human manipulation such as chemical, physical, stress-induced, or other means, has undergone mutation and selection; or that an exogenous nucleic acid has actually been introduced to the cell or entity through any standard means, such as transfection; such that the cell or entity has acquired a new characteristic, phenotype, genotype, and/or gene expression product, including but not limited to a gene marker, a gene product, and/or a mRNA, to endow the original cell or entity, at a genetic level, with a function, characteristic, or genetic element not present in non-genetically engineered, non-selected counterpart cells or entities.
  • Fibroblast cells generally, produce a number of extracellular matrix proteins, primarily collagen. There are several types of collagens produced by fibroblasts, however, type I collagen is the most prevalent in vivo.
  • Human fibroblast cell strains can be derived from a number of sources, including, but not limited to neonate male foreskin, dermis, tendon, lung, umbilical cords, cartilage, urethra, corneal stroma, oral mucosa, and intestine.
  • the human cells may include but need not be limited to fibroblasts, but may include: smooth muscle cells, chondrocytes and other connective tissue cells of mesenchymal origin. It is preferred, but not required, that the origin of the matrix-producing cell used in the production of a tissue construct be derived from a tissue type that it is to resemble or mimic after employing the culturing methods of the invention. For instance, where a skin-construct is produced, the matrix- producing cell is a fibroblast, preferably of dermal origin. In another preferred embodiment, fibroblasts isolated by microdissection from the dermal papilla of hair follicles can be used to produce the matrix alone or in association with other fibroblasts.
  • the matrix-producing cell is derived from corneal stroma.
  • Cell donors may vary in development and age.
  • Cells may be derived from donor tissues of embryos, neonates, or older individuals including adults.
  • adult stem cells e.g. bone marrow derived stem cells
  • human cells are preferred for use in the invention, the cells to be used in the method of are not limited to cells from human sources.
  • Cells from other mammalian species including, but not limited to, equine, canine, porcine, bovine, and ovine sources; or rodent species such as mouse or rat may be used.
  • cells that are spontaneously, chemically or virally transfected or recombinant cells or genetically engineered cells may also be used in this invention.
  • chimeric mixtures of normal cells from two or more sources; mixtures of normal and genetically modified or transfected cells; or mixtures of cells of two or more species or tissue sources may be used.
  • the matrix-producing cell is cultured in a vessel suitable for animal cell or tissue culture, such as a culture dish, flask, or roller-bottle, which allows for the formation of a three- dimensional tissue-like structure.
  • a vessel suitable for animal cell or tissue culture such as a culture dish, flask, or roller-bottle, which allows for the formation of a three- dimensional tissue-like structure.
  • Suitable cell growth surfaces on which the cells can be grown can be any biologically compatible material to which the cells can adhere and provide an anchoring means for the cell-matrix construct to form.
  • Materials such as glass; stainless steel; polymers, including polycarbonate, polystyrene, polyvinyl chloride, polyvinylidene, polydimethylsiloxane, fluoropolymers, and fluorinated ethylene propylene; and silicon substrates, including fused silica, polysilicone, or silicon crystals may be used as a cell growth surfaces.
  • tissue construct of the invention may be grown on a solid cell growth surface, a cell growth surface with pores that communicate both top and bottom surfaces of the membrane to allow bilateral contact of the medium to the developing tissue construct or for contact from only below the culture is preferred. Bilateral contact allows medium to contact both the top and bottom surfaces of the developing construct for maximal surface area exposure to the nutrients contained in the medium. Medium may also contact only the bottom of the forming cultured tissue construct so that the top surface may be exposed to air, as in the development of a cultured skin construct.
  • the preferred culture vessel is one that utilizes a carrier insert, a culture-treated permeable member such as a porous membrane that is suspended in the culture vessel containing medium.
  • the membrane is secured to one end of a tubular member or framework that is inserted within and interfaces with a base, such as a petri or culture dish that can be covered with a lid.
  • a base such as a petri or culture dish that can be covered with a lid.
  • the tissue-construct is produced on one surface of the membrane, preferably the top, upwardly facing surface and the culture is contacted by cell media on both top and bottom surfaces.
  • the pores in the growth surface allow for the passage of culture media for providing nutrients to the underside of the culture through the membrane, thus allowing the cells to be fed bilaterally or solely from the bottom side.
  • a preferred pore size is one that is small enough that it does not allow for the growth of cells through the membrane, yet large enough to allow for free passage of nutrients contained in culture medium to the bottom surface of the cell-matrix construct, such as by capillary action.
  • Preferred pore sizes are about less than 3 microns but range between about 0.1 microns to about 3 microns, more preferably between about 0.2 microns to about 1 micron and most preferably about 0.4 micron to about 0.6 micron sized pores are employed.
  • the most preferred material is polycarbonate having a pore size is between about 0.4 to about 0.6 microns.
  • the maximum pore size depends not only on the size of the cell but also the ability of the cell to alter its shape and pass through the membrane. It is important that the tissue-like construct adheres to the surface but does not incorporate or envelop the substrate so it is removable from it such as by peeling with minimal force.
  • the size and shape of the tissue construct formed is dictated by the size of the vessel surface or membrane on which it grown. Substrates may be round or angular or shaped with rounded corner angles, or irregularly shaped. Substrates may also be flat or contoured as a mold to produce a shaped construct to interface with a wound or mimic the physical structure of native tissue.
  • tissue construct is finally formed, whether it is a single layer cell-matrix construct or a bi-layer construct, it is removed by peeling from the membrane substrate before grafting to a patient.
  • the system for the production of the cell-matrix layer may be either static or may employ a perfusion means to the culture media.
  • the culture medium In the static system, the culture medium is still and relatively motionless as contrasted to the perfusion system where the medium is in motion.
  • the perfusion of medium affects the viability of the cells and augments the development of the matrix layer.
  • Perfusion means include, but are not limited to: using a magnetic stir bar or motorized impeller in the culture dish subjacent (below) or adjacent to the substrate carrier containing the culture membrane to stir the medium; pumping medium within or through the culture dish or chamber; gently agitating the culture dish on a shaking or rotating platform; or rolling, if produced in a roller bottle.
  • Culture media formulations suitable for use in the present invention are selected based on the cell types to be cultured and the extracellular matrix to be produced. The culture medium that is used and the specific culturing conditions needed to promote cell growth, matrix synthesis, and viability will depend on the type of cell being grown.
  • chemically defined culture media is preferred, that is, media free of undefined animal organ or tissue extracts, for example, serum, pituitary extract, hypothalamic extract, placental extract, or embryonic extract or proteins and factors secreted by feeder cells.
  • the media are free of undefined components and defined biological components derived from non-human sources.
  • the resultant tissue construct is a defined human tissue construct.
  • Synthetic functional equivalents may also be added to supplement chemically defined media within the purview of the definition of chemically defined for use in the most preferred fabrication method.
  • suitable natural human, human recombinant, or synthetic equivalents to commonly known animal components to supplement the culture media of the invention without undue investigation or experimentation.
  • Culture medium is comprised of a nutrient base usually further supplemented with other components.
  • the skilled scientist can determine appropriate nutrient bases in the art of animal cell culture with reasonable expectations for successfully producing a tissue construct of the invention.
  • Many commercially available nutrient sources are useful on the practice of the present invention. These include commercially available nutrient sources which supply inorganic salts, an energy source, amino acids, and B-vitamins such as Dulbecco's Modified Eagle's Medium (DMEM); Minimal Essential Medium (MEM); M199; RPMI 1640; Iscove's Modified Dulbecco's Medium (EDMEM).
  • DMEM Dulbecco's Modified Eagle's Medium
  • MEM Minimal Essential Medium
  • M199 require additional supplementation with phospholipid precursors and nonessential amino acids.
  • vitamin-rich mixtures that supply additional amino acids, nucleic acids, enzyme cofactors, phospholipid precursors, and inorganic salts include Ham's F-12, Ham's F-10, NCTC 109, and NCTC 135. Albeit in varying concentrations, all basal media provide a basic nutrient source for cells in the form of glucose, amino acids, vitamins, and inorganic ions, together with other basic media components.
  • the most preferred base medium of the invention comprises a nutrient base of either calcium-free or low calcium Dulbecco's Modified Eagle's Medium (DMEM), or, alternatively , DMEM and Ham's F-12 between a 3-to-1 ratio to a 1 -to-3 ratio, respectively.
  • DMEM Dulbecco's Modified Eagle's Medium
  • the base medium is supplemented with components such as amino acids, growth factors, and hormones.
  • culture media for the culture of cells of the invention are described in United States Patent No. 5,712,163 and in International PCT Publication No. WO 95/31473 the disclosures of which are incorporated herein by reference.
  • Other media are known in the art such as those disclosed in Ham and McKeehan, Methods in Enzymology,
  • the base medium is supplemented with the following components known to the skilled artisan in animal cell culture: insulin, transferrin, triiodothyronine (T3), and either or both ethanolamine and o- phosphoryl-ethanolamine, wherein concentrations and substitutions for the supplements may be determined by the skilled artisan.
  • Insulin is a polypeptide hormone that promotes the uptake of glucose and amino acids to provide long term benefits over multiple passages. Supplementation of insulin or insulin-like growth factor (IGF) is necessary for long term culture as there will be eventual depletion of the cells' ability to uptake glucose and amino acids and possible degradation of the cell phenotype. Insulin may be derived from either animal, for example bovine, human sources, or by recombinant means as human recombinant insulin. Therefore, a human insulin would qualify as a chemically defined component not derived from a non-human biological source. Insulin supplementation is advisable for serial cultivation and is provided to the media at a wide range of concentrations.
  • IGF insulin-like growth factor
  • a preferred concentration range is between about 0.1 ⁇ g/ml to about 500 ⁇ g/ml, more preferably at about 5 ⁇ g/ml to about 400 ⁇ g/ml, and most preferably at about 375 ⁇ g/ml.
  • Appropriate concentrations for the supplementation of insulin-like growth factor, such as IGF-1 or IGF-2, may be easily determined by one of skill in the art for the cell types chosen for culture.
  • Transferrin is in the medium for iron transport regulation. Iron is an essential trace element found in serum. As iron can be toxic to cells in its free form, in serum it is supplied to cells bound to transferrin at a concentration range of preferably between about 0.05 to about 50 ⁇ g/ml, more preferably at about 5 ⁇ g/ml.
  • Triiodothyronine (T3) is a basic component and is the active form of thyroid hormone that is included in the medium to maintain rates of cell metabolism. Truodothyronine is supplemented to the medium at a concentration range between about 0 to about 400 pM, more preferably between about 2 to about 200 pM and most preferably at about 20 pM.
  • Either or both ethanolamine and o-phosphoryl-ethanolamine, which are phospholipids, are added whose function is an important precursor in the inositol pathway and fatty acid metabolism. Supplementation of lipids that are normally found in serum is necessary in a serum-free medium. Ethanolamine and o-phosphoryl-ethanolamine are provided to media at a concentration range between about 10 '6 to about 10 '2 M, more preferably at about 1 x 10 '4 M.
  • the base medium is additionally supplemented with other components to induce synthesis or differentiation or to improve cell growth such as hydrocortisone, selenium, and L-glutamine.
  • Hydrocortisone has been shown in keratinocyte culture to promote keratinocyte phenotype and therefore enhance differentiated characteristics such as involucrin and keratinocyte transglutaminase content (Rubin et al., J. Cell PhysioL, 138:208-214 (1986)). Therefore, hydrocortisone is a desirable additive in instances where these characteristics are beneficial such as in the formation of keratinocyte sheet grafts or skin constructs. Hydrocortisone may be provided at a concentration range of about 0.01 ug/ml to about 4.0 ⁇ g/ml, most preferably between about 0.4 ⁇ g/ml to 16 ug/ml.
  • Selenium is added to serum-free media to resupplement the trace elements of selenium normally provided by serum.
  • Selenium may be provided at a concentration range of about 10 ⁇ 9 M to about 10 ⁇ 7 M; most preferably at about 5.3 x 10 "8 M.
  • L-glutamine is present in some nutrient bases and may be added in cases where there is none or insufficient amounts present. L-glutamine may also be provided in stable form such as that sold under the mark, GlutaMAX-1 TM (Gibco BRL, Grand Island,
  • GlutaMAX-1 TM is the stable dipeptide form of L-alanyl-L-glutamine and may be used interchangeably with L-glutamine and is provided in equimolar concentrations as a substitute to L-glutamine.
  • the dipeptide provides stability to L-glutamine from degradation over time in storage and during incubation that can lead to uncertainty in the effective concentration of L-glutamine in medium.
  • the base medium is supplemented with preferably between about 1 mM to about 6 mM, more preferably between about 2 mM to about 5 mM, and most preferably 4 mM L-glutamine or GlutaMAX-1 TM.
  • EGF epidermal growth factor
  • EGF in native form or recombinant form may be used.
  • Human forms, native or recombinant, of EGF are preferred for use in the medium when fabricating a skin equivalent containing no non- human biological components.
  • EGF is an optional component and may be provided at a concentration between about 1 to 15 ng/mL, more preferably between about 5 to 10 ng/mL.
  • the medium described above is typically prepared as set forth below. However, it should be understood that the components of the present invention may be prepared and assembled using conventional methodology compatible with their physical properties. It is well known in the art to substitute certain components with an appropriate analogous or functionally equivalent acting agent for the purposes of availability or economy and arrive at a similar result. Naturally occurring growth factors may be substituted with recombinant or synthetic growth factors that have similar qualities and results when used in the performance of the invention.
  • Media in accordance with the present invention are sterile. Sterile components are bought sterile or rendered sterile by conventional procedures, such as filtration, after preparation. Proper aseptic procedures were used throughout the following Examples. DMEM and F-12 are first combined and the individual components are then added to complete the medium.
  • transferrin is dissolved in water.
  • the three are then mixed and diluted in water to a 500X concentration.
  • Ethanolamine and o-phosphoryl-ethanolamine are dissolved in water to 500X concentration and are filter sterilized.
  • Progesterone is dissolved in absolute ethanol and diluted with water.
  • Hydrocortisone is dissolved in absolute ethanol and diluted in phosphate buffered saline (PBS).
  • Selenium is dissolved in water to 500X concentration and filter sterilized.
  • EGF is purchased sterile and is dissolved in PBS. Adenine is difficult to dissolve but may be dissolved by any number of methods known to those skilled in the art.
  • Serum albumin may be added to certain components in order to stabilize them in solution and are presently derived from either human or animal sources.
  • human serum albumin (HSA) or bovine serum albumin (BSA) may be added for prolonged storage to maintain the activity of the progesterone and EGF stock solutions.
  • the medium can be either used immediately after preparation or, stored at 4 0 C. If stored, EGF should not be added until the time of use.
  • the medium is supplemented with additional agents that promote matrix synthesis and deposition by the cells.
  • These supplemental agents are cell-compatible, defined to a high degree of purity and are free of contaminants.
  • the medium used to produce the cell-matrix is termed "matrix production medium”.
  • the base medium is supplemented with an ascorbate derivative such as sodium ascorbate, ascorbic acid, or one of its more chemically stable derivatives such as L-ascorbic acid phosphate magnesium salt n-hydrate.
  • Ascorbate is added to promote hydroxylation of proline and secretion of procollagen, a soluble precursor to deposited collagen molecules.
  • Ascorbate has also been shown to be an important cofactor for post-translational processing of other enzymes as well as an upregulator of type I and type III collagen synthesis.
  • proline and glycine are basic amino acids that make up the structure of collagen.
  • the matrix-production medium is optionally supplemented with a neutral polymer.
  • the cell-matrix constructs of the invention may be produced without a neutral polymer, but again not wishing to be bound by theory, its presence in the matrix production medium may collagen processing and deposition more consistently between samples.
  • One preferred neutral polymer is polyethylene glycol (PEG), which has been shown to promote in vitro processing of the soluble precursor procollagen produced by the cultured cells to matrix deposited collagen.
  • Tissue culture grade PEG within the range between about 1000 to about 4000 MW (molecular weight), more preferably between about 3400 to about 3700
  • Preferred PEG concentrations are for use in the method may be at concentrations at about 5% w/v or less, preferably about 0.01 % w/v to about 0.5% w/v, more preferably between about 0.025% w/v to about 0.2% w/v, most preferably about 0.05% w/v.
  • Other culture grade neutral polymers such as dextran, preferably dextran T-40, or polyvinylpyrrolidone (PVP), preferably in the range of 30,000-40,000 MW, may also be used at concentrations at about 5% w/v or less, preferably between about 0.01% w/v to about 0.5% w/v, more preferably between about 0.025% w/v to about 0.2% w/v, most preferably about 0.05% w/v.
  • Other cell culture grade and cell-compatible agents that enhance collagen processing and deposition may be ascertained by the skilled routineer in the art of mammalian cell culture.
  • a preferred matrix production medium formulation comprises: a base 3:1 mixture of Dulbecco's Modified Eagle's Medium (DMEM) (high glucose formulation, without L-glutamine) and Hams F-12 medium supplemented with either 4 mM L-glutamine or equivalent, 5 ng/ml epidermal growth factor, 0.4 ⁇ g/ml hydrocortisone, 1 x 10 '4 M ethanolamine, 1 x 10 '4 M o-phosphoryl-ethanolamine, 5 ⁇ g/ml insulin, 5 ⁇ g/ml transferrin, 20 pM triiodothyronine, 6.78 ng/ml selenium, 50 ng/ml L-ascorbic acid, 0.2 ⁇ g/ml
  • DMEM Dulbecco's Modified Eagle's Medium
  • Hams F-12 medium supplemented with either 4 mM L-glutamine or equivalent, 5 ng/ml epidermal growth factor, 0.4 ⁇ g/ml hydrocor
  • pharmacological agents may be added to the culture to alter the nature, amount, or type of the extracellular matrix secreted.
  • agents may include polypeptide growth factors, transcription factors or inorganic salts to up- regulate collagen transcription.
  • polypeptide growth factors include transforming growth factor-beta 1 (TGF- ⁇ 1 ) and tissue-plasmmogen activator (TPA), both of which are known to upregulate collagen synthesis.
  • TGF- ⁇ 1 transforming growth factor-beta 1
  • TPA tissue-plasmmogen activator
  • An example of an inorganic salt that stimulates collagen production is cerium. Shivakumar et al., Journal of Molecular and Cellular Cardiology 24:775-780 (1992).
  • the cultures are maintained in an incubator to ensure sufficient environmental conditions of controlled temperature, humidity, and gas mixture for the culture of cells.
  • Preferred conditions are between about 34 0 C to about 38 0 C, more preferably 37 ⁇ 1 0 C with an atmosphere between about 5-10 ⁇ 1 % CO 2 and a relative humidity (Rh) between about 80-
  • cells are harvested and seeded onto a suitable culture surface and cultured under appropriate growth conditions to form a confluent sheet of cells.
  • the cells are seeded on a porous membrane that is submerged to allow medium contact from below the culture through the pores and directly above.
  • cells are suspended in either base or growth media and are seeded on the cell culture surface at a density between about 1 x 10 5 cells/cm 2 to about 6.6 x 10 5 cells/cm 2 , more preferably between about 3 x 10 5 cells/cm 2 to about 6.6 x
  • Cultures are cultured in growth medium to establish the culture and are cultured to between about 80% to 100% confluence at which time they are induced chemically by changing the medium to matrix production medium in order to upregulate the synthesis and secretion of extracellular matrix.
  • cells are seeded directly in production media to eliminate the need to change from the basic media to the production media but it is a method that requires higher seeding densities.
  • the cells organize the secreted matrix molecules to form a three dimensional tissue-like structure but do not exhibit significant contractile forces to cause the forming cell-matrix construct to contract and peel itself from the culture substrate.
  • Media exchanges are made every two to three days with fresh matrix production medium and with time, the secreted matrix increases in thickness and organization. The time necessary for creating a cell-matrix construct is dependent on the ability of the initial seeding density, the cell type, the age of the cell line, and the ability of the cell line to synthesize and secrete matrix.
  • the constructs of the invention When fully formed, the constructs of the invention have bulk thickness due to the fibrous matrix produced and organized by the cells; they are not ordinary confluent or overly confluent cell cultures where the cells may be loosely adherent to each other.
  • the fibrous quality gives the constructs cohesive tissue-like properties unlike ordinary cultures because they resist physical damage, such as tearing or cracking, with routine handling in a clinical setting.
  • the cells In the fabrication of a cultured dermal construct, the cells will form an organized matrix around themselves on the cell culture surface preferably at least about 30 microns in thickness or more, more preferably between about 60 to about 120 microns thick across the surface of the membrane; however, thicknesses have been obtained in excess of 120 microns and are suitable for use in testing or clinical applications where such greater thicknesses are needed.
  • mixed cell populations of two or more cell types may be cultured together during the formation of a tissue construct of the invention provided that at least one of the cell types used is capable of synthesizing extracellular matrix.
  • the second cell type may be one needed to perform other tissue functions or to develop particular structural features of the tissue construct.
  • the production of the matrix in vitro in accordance with the present invention has shown to mimic several of the processes exhibited in production of matrix as well as repair of matrix in vivo.
  • extracellular matrix extracts are used for the induction of organ/tissue specific cell phenotypes from cultured bone marrow-derived mesenchymal stem cells in vitro. These extracts contain all the growth factors and cytokines present in the extracellular matrix necessary for specific organ differentiation.
  • the present example is based on the production and use of cartilage extracellular matrix extracts.
  • the method of the present invention requires the addition of an extracellular matrix extract (including soluble factors) closely resembling the matrix of the tissue cells into which the stem cells should differentiate.
  • Such inducing extracts may be prepared in various ways. In particular they may be derived directly from the organ/tissue of interest, either from a human or an animal, which matrix closely resembles that of a human. Alternatively, the extracts may be made synthetically by blending commercially available growth factors, differentiation factor etc in order to produce an extract closely resembling the corresponding in vivo extracellular matrix.
  • the extracellular matrix extracts are organ-derived extracts on equine basis (supplied by Ossacur, Oberstenfeld, Germany). These organ-derived extracts are sterile acellular lyophilizates extracted from cartilage. In order to demonstrate the applicability of these extracts to induce differentiation of the stem cells and extracellular matrix changes in cell morphology, proliferation rate, expression of typical chondrogenic differentiation markers have been examined. Most importantly, the identification of SOX9 (Crombrugghe et al. 2001 and Goldring et al. 2006), a major nuclear transcription factor expressed in cells undergoing condensation and required for the expression of aggrecan cartilage-specific matrix protein (Lin et al. 2005), and the presence of glycosaminoglycans
  • Bone marrow aspirate was then subjected to hypoaque density gradient technique by centrifugation at 1400 rpm for 30 minutes at room temperature. Buffy coat, containing nucleated cells, was isolated and re-suspended in 10 ml culture medium compromising
  • Dulbecco's modified Eagles medium 1000mg glucose/L, L-glutamine, NaHCO and pyridoxine.HCI
  • penicillin / streptomycin Sigma-Aldhch Co Ltd, Irvine UK
  • 10% fetal bovine serum GEBCO-BRL
  • Supernatant was the removed and the cell pallet was re-suspended in medium by 3 successive aspirations and expulsions through a 23-gauge needle attached to a 5 ml syringe.
  • Cells were then plated in a 100 mm petri dish and incubated at 37 0 C in a 95% air and 5% carbon dioxide at 100% humidity.
  • Stro-1 cell surface marker was used to confirm that cell cultures comprised a multipotential bone marrow stem cell population. Cells expressing the Stro-1 cell surface
  • Stro-1 monoclonal antibody R&D systems, Minneapolis, MN
  • Primary antibody was then removed from the wells and cells were washed three times with phosphate buffered saline, before incubation for 1 hour with anti mouse IgM FITC-conjugated (Molecular Probes) at a concentration of 1 :1000 for 1 hour .
  • Secondary antibody was removed and cells were further washed 3 times with PBS. A negative control was performed with the secondary antibody. After the incubation period cells were washed, mounted onto slides and observed using a fluorescent microscope.
  • H&E H&E stain was used to assess both the morphology of the cells and the presence of calcium deposits in the culture. 14 days after treatment, cells seeded on coverslips were fixed with 70% ethanol, hydrated with distilled water for 3 minutes, placed in hematoxylin for 4 minutes then washed once with tap water for 3 minutes. Next, the coverslips were dipped in eosin for 45 seconds followed by a dehydration step consisting of 95% ethanol for 3 minutes (twice) then 100% ethanol for 3 minutes. Cells were then placed in xylol for 3 minutes then mounted. Cells were visualized using light microscopy.
  • Toluidine blue is a monocationic dye which has been used for the staining of proteoglycans (PGs) in tissue for histochemical studies. Cells were hydrated using water then incubated 2-
  • G Quantification of Chondroitin Sulfate Alcian blue is a tetracationic dye which has been used by various investigators for the quantification of microgram quantities of nonradioactive GAG and PG.
  • GAGs and PGs were precipitated specifically by addition of Alcian blue under conditions of low pH and of critical electrolyte concentrations, which prevent interaction of the dye with other anionic macromolecules such as nucleic acids or proteins.
  • Cells were washed with PBS then incubated in Alcian Blue stain (1% Alcian Blue in 3% acetic acid) for 20 min. Cells were destained by three rinses in 3% acetic acid and then washed once in water. To quantify stain incorporated, cells were solubilized in 1 % SDS by shaking for 30 min at room temperature and then heating to 90 0 C for 1 h. Absorbance was detected at 595 nm.
  • the pellet was washed three times with phosphate buffered saline (PBS), and then centrifuged for 2 minutes at 12500 rpm. The supernatant was removed and the pellet was re-suspend in 40 ⁇ l 2X sample buffer 5% ⁇ -mercaptoethanol. The samples were boiled for 5 minutes and ran on SDS-PAGE gel. Note: samples were processed in parallel but without the addition of TGF- ⁇ 1 antibody.
  • PBS phosphate buffered saline
  • lysis buffer consisting of 0.125M Tris-HCI (pH 6.8), 2% SDS, 5 % B-mercaptoethanol, and 10% glycerol.
  • Samples were loaded onto a 12% SDS-polyacrylamide gel, subjected to electrophoresis, and transferred to PVDF membrane. After blocking the membranes in 5% skim milk in Tris-buffered saline containing 0.05% Tween-20, the blots were incubated with specific antibodies (anti-TGF ⁇ 1 antibody). The blots were washed, and protein bands were visualized by chemiluminescence. Proteins were coomassie stained to ensure both equal loading and quality of the proteins extracted. GAPDH was also used for equal loading.
  • Osteopontin (OPN) secretion was analyzed using a human OPN TiterZyme enzyme-linked immunosorbent assay (ELISA) Kit (Assay designs). Cell supernatant was collected at day 14 post treatment. A volume of 100 ⁇ l assay buffer provided in the kit and 100 ⁇ l of the samples or standard solution were added into a microtiter plate. A series of standard solutions were prepared by diluting the stock solution containing recombinant human OPN (16ng/ml). The micotiter plate with the samples was incubated at room temperature for 1 h, the solutions in the wells were aspirated and the wells were rinsed by adding 400 ⁇ l of washing buffer 4 times.
  • ELISA enzyme-linked immunosorbent assay
  • NucleoSpin RNA Il from Macherey-Nagel.
  • NM000088 Collagen type I
  • SOX9 NM_000346
  • RNA quality was verified by RNA quality and ensure equal loading of the samples.
  • RT-PCR Real-time reverse transcription polymerase chain reaction
  • the real-time PCR conditions consisted of a reverse transcription step for 20 minutes at 61 9 C to generate single-stranded cDNA from mRNA followed by a reverse transcriptase inactivation and initial denaturation for 30 seconds at 95 9 C. This was followed by 40 cycles of amplification consisting of 95 9 C for 1 second, 58 9 C for 15 seconds and 72 9 C for 13 seconds. A final cooling step at 40 9 C for 30 seconds was then performed. The concentration of GAPDH was used to control for input of RNA and to normalize all genes tested from the same cDNA sample. RESULTS
  • BMMSC isolation, characterization and morphology Approximately 2.5 - 4 ml of bone marrow aspirates were obtained from patients after obtaining their informed consent. After ficoll, the yield of nucleated cells extracted was 1 10x10 4 cells per 1 ml of bone marrow aspirate. Cells were then plated, replenished with new culture medium every 3 days. Cultured BMMSC were observed by light microscopy and morphology was documented using digital photography. After 7-10 days in culture, cells started to adhere to the plastic of the culture vessel forming colonies. At day 14, these cells were trypsinized and seeded into new culture vessels, a process known as passaging. With repeated passaging, the cells exhibited a characteristic elongated morphology. The identity of these cells as BMMSC was conformed by staining for Stro-1 , an antigen marker specific for BMMSC. These cells positively stained for Stro-1 .
  • TGF- ⁇ 1 is one of the growth factors known in the enhancement of chondrocytic differentiation of BMMSC. TGF- ⁇ 1 was identified in of joint and meniscus matrix extracts using an immunoprecipitation technique.
  • Sox9 being a key transcription factor that plays a role in chondrogenesis
  • Joint and meniscus extracts induce chondrogenic differentiation of BMMSCs through the upregulation of Sox9 mRNA (Fig 2).
  • Collagen type I known to be upregulated during osteogenic differentiation, was not detected (Fig 3). This data suggests the onset of chondrogenesis.
  • Aggrecan a large chondroitin sulfate proteoglycan, is one of the major structural components in cartilage matrix. It binds to hyaluronan and Link proteins to form huge aggregates.
  • the aggrecan molecule is composed of a core protein and glycosaminoglycans, mostly chondroitin sulfate.
  • Aggrecan was detectable at the protein level at day 14 post treatment using mouse anti-aggrecan antibody.
  • the expression of aggrecan was highly distributed and localized in chondrocytic differentiating human mesenchymal stem cells as compared to untreated cells (Fig 4). In parallel, staining with the secondary antibody was performed to ensure the specific binding of anti-aggrecan antibody (data not shown).
  • Alcian blue and Toluidine blue dyes form complexes with anionic glycoconjugates (AG) such as proteoglycans (PG) and glycosaminoglycans (GAG).
  • AG anionic glycoconjugates
  • PG proteoglycans
  • GAG glycosaminoglycans
  • alcian blue was used to quantitatively measure the amount of chondroitin sulfate in cell lysates.
  • Our results showed an increase in the amount of chondroitin sulfate in both joint and meniscus extracts as compared to the control untreated cells. It was noted that cells treated with bovine joint extract contained more chondroitin sulfate than that of bovine meniscus extract.
  • the addition of 10ng/ml TGF- ⁇ 1 to the culture was conducted in parallel manifesting an increase in the level of chondroitin sulfate.
  • toluidine blue staining is used to histologically detect glycosaminoglycans (GAGs) in BMMSC cultures in the presence of joint and meniscus extract. Results show a clear blue cytoplasm with violet/red purple granules indicative of the presence of glycosaminoglycans GAGs.
  • Cartilage derived matrix extracts do not induce osteocytic differentiation of BMMSC in vitro
  • Osteopontin is known to be secreted and expressed as a result of BMMSC differentiation into osteocytes and not chondrocytes.
  • OPN osteopontin
  • cellular supernatant was collected from all cultures. Levels of OPN were same as that of the control untreated cells.
  • Colloss and Colloss E previously used in differentiating BMMSC into osteocytes (El-Sabban et al. 2007), treated cultures showed high levels of secreted OPN in their supernatant.
  • 10ng/ml of BMP-2 and BMP-7 (OP-1 ) recombinant proteins were used as a positive control for the induction of BMMSC differentiation into osteocytes.
  • osteopontin was also secreted.
  • cartilage derived joint and meniscus extracts did not induce the secretion of osteopontin into the supernatant.
  • Treatement of BMMSC with cartilage derived matrix extracts resulted in low levels of collagen type I equivalent to that of control untreated cells.
  • a significant increase in collagen type I expression was detected in BMMSC treated with bone derived matrix extracts (Colloss and Colloss-E) as compared to untreated BMMSC. This suggests the exclusive expression of collagen type I in BMSC differentiation into osteocytes rather than chondrocytes.
  • the cultivation broth comprising differentiated human cells and extracellular matrix
  • the supernatant constitutes the extracellular matrix extract of the present invention.
  • This human based extract has a composition which more closely resembles the extracellular matrix of human cartilage tissue than the equine base extract does. Accordingly, the present invention provides a more perfectly resembling extracellular matrix extract than the prior art envisages.
  • the present invention also contemplates more simple extracellular extracts resembling those derived from animals or humans; instead of deriving the extract directly from the tissue of interest it is possible to manufacture a more simple but still useful extract for the stimulation of stem cells to produce extracellular matrix.
  • the present inventors have been able to stimulate stem cells to produce extracellular matrix of bone, cartilage, bone marrow, pancreas and liver by simply mixing human growth and differentiation factors in appropriate ratios in accordance with Table 1.
  • Bone marrow-derived mesenchymal stem cells are infrequent multipotent cells that can differentiate into multiple lineages, such as bone, cartilage and muscle cells among others. Hence, these cells are of potential clinical importance for the repair of damaged tissues.
  • the local microenvironment in vivo is critical to support the desired differentiation of stem cells or to sustain the phenotype of the stem cell-derived in vitro differentiated cells.
  • This local microenvironment comprises a physical support supplied by the organ matrix as well as tissue-specific cytokines. Bone matrix contains collagen type 1 and many of the growth factors involved in the cascade of bone formation. Consequently, when bone marrow is used to enrich orthopedic grafting matrices, it almost invariably produces faster and more consistent defect healing compared with bone marrow or the carrier matrix alone.
  • BMEs xenogenic bone matrix extracts
  • Bovine and equine bone protein extracts were supplied by Ossacur, AG (Oberstenfeld, Germany). These extracts were prepared, aseptically, from cortical diaphysis of long bones from disease-free, young ( ⁇ 12 months old) calves and foals of closed herds. Briefly, bones were pulverized and delipidated with acetone for 60 min at 4"C. The resulting bone particles were demineralized in 0.6N hydrochloric acid for 60 min at 4 0 C. Particles were then washed in deionized water, extracted with 4M guanidine hydrochloride and ultrafiltered using 3 K nominal molecular weight cut-off membranes. Colloss and Colloss-E are normally supplied as sterile lyophilizates that comprise a suspension when reconstituted. Hence, Colloss and Colloss-E dispersion in water, an intermediate stage from the manufacturing procedure, was used for this study.
  • Human BMMSCs were isolated from 3 ⁇ ml of bone marrow aspirates using Ficoll density gradient (Ficoll-PaqueTM Plus, Amersham Bioscience, UK). Cells were cultured in Dulbecco's modified Eagles medium (1000 mg/l-glucose/L, L-glutamine and sodium pyruvate; Sigma Aldrich, USA), with 1 % penicllin/streptomycin, fetal bovine serum (Gibco BRL, USA) at 37 0 C in 95% air with 5% carbon dioxide at 100% humidity. Medium was replenished every 2 days. Confluent cultures were passaged by trypsinization and gentle scraping. Cells between passage five and seven were used for all experiments.
  • BMMSCs were treated for 7 or 14 days with medium containing 1 :50 Colloss or Colloss-E (28 and 32 ⁇ g/ml, respectively).
  • Alkaline phosphatase protein levels in total cell lysates were evaluated by western blot using specific antibodies (Abeam, UK). Alkaline phosphatase enzyme activity was assessed in medium-free cell lysates as described by Oreffo and colleagues using 5 mM of p-nitrophenyl phosphate (Amresco, USA) as a substrate in 2- amino-2-methyl-1 propanol (Sigma) alkaline buffer solution.
  • BMMSCs were treated for 7, 14 and 30 days with medium containing Colloss or Colloss-E
  • Osteopontin levels were determined in cell-free supernatant using a human OPN TiterZyme ELISA kit (Assay designs, USA) as recommended by the manufacturer.
  • BMMSCs were treated for 15 days with medium containing 1 :50 Colloss or Colloss-E (28 and 32 ⁇ g/ml, respectively). Cultures were stained with hematoxylin and eosin (H&E) and calcium deposits were counted.
  • H&E hematoxylin and eosin
  • Protein levels of various osteogenic cytokines in Colloss-E were determined by ELISA (antibodies generated against human factors), according to the supplier's instructions. Levels of BMP-2, TFG- ⁇ i and insulin-like growth factor (IGF)-I were measured using commercial ELISA kits from R&D Systems, whereas levels of BMP-7 were measured using commercial ELISA kits from RayBiotech.
  • Bovine & equine BMEs induce morphological changes of BMMSC
  • Bovine & equine BMEs induce osteoblastic differentiation of BMMSCs
  • Colloss or Colloss-E induce osteoblastic differentiation of BMMSCs
  • treated cells were assessed for the presence of osteoblast-specific markers, such as alkaline phosphatase activity, osteopontin secretion and calcium deposits.
  • Ostblast-specific markers such as alkaline phosphatase activity, osteopontin secretion and calcium deposits.
  • Both Colloss and Colloss-E induced a sharp increase in alkaline phosphatase protein level at day 14 ( Figure 8A). This was accompanied by a significant increase in alkaline phosphatase enzyme activity in medium-free cell extracts at day 14 ( Figure 8B).
  • Colloss and Colloss-E were devoid of alkaline phosphatase activity (data not shown).
  • Colloss-E ® contains osteogenic growth factors
  • Colloss-E contains significant amounts of BMP-2 (2.6 ⁇ 0.2 mg/g), BMP-7 (3.8 ⁇ 2.7 mg/g), TGF- ⁇ 1 (55 ⁇ 1 1 mg/g) and IGF-1 (2.9 ⁇ 0.8 mg/g).
  • Colloss had opposite effects depending on the differentiated state of the cells. Whereas, Colloss increased proliferation and decreased alkaline phosphatase activity of undifferentiated hMSC-TERT cells, it diminished proliferation and increased alkaline phosphatase activity and collagen synthesis of osteoblastic differentiated hMSC-TERT cells.
  • our report is the first to demonstrate that Colloss and Colloss-E induce osteoblastic differentiation of in wf/O-expanded human BMMSCs.
  • xenogenic BME-induced osteoblastic differentiation of BMMSCs remains to be understood.
  • Normal in vivo cell differentiation is coordinated by both a structural scaffold and soluble cytokines, both provided by the microenvironment of target organs.
  • organ-specific extracts to supply stem cells with a milieu that mimics the in vivo microenvironment is an attractive alternative to the use of a defined combination of cytokines and growth factors.
  • These extracts provide both the physical and biochemical signals in relative physiologic proportions.
  • xenogenic BMEs consist of collagens as well as noncollagenous-associated proteins.
  • Colloss-E contains significant amounts of osteogenic growth factors, such as BMP-2, BMP-7, TGF- ⁇ 1 and IGF- 1.
  • Colloss and Colloss-E induce osteoblastic differentiation of in vitro expanded BMMSCs, which has far-reaching applications in the treatment of bone defects.
  • These extracts can be co-administered with autologous BMMSC in the area of the bone defect, to enhance their osteoblastic differentiation.
  • Colloss and Colloss-E can be used as an ex vivo tissue engineering scaffold to induce in vitro osteoblastic differentiation of expanded BMMSCs, which can then be applied in vivo. This provides a safe, inexpensive and readily available source of osteoinductive graft materials.
  • the present example illustrates the applicability of the present invention to differentiate human BMMSCs into hepatocytes thereby producing valuable human based ECM and soluble factors.
  • ECM and soluble factors are obtained by removing the differentiated cells leaving ECM and soluble factors; commonly denoted ECM extract.
  • This extract may then be stored and later used as differentiation medium for the differentiation of BMMSCs into hepatocytes (ex vivo or in vivo).
  • the differentiation of the human BMMSCs into hepatocytes is visualized in Figure 13, whereas the biochemical changes associated therewith are shown in Figures 1 1 and 12.
  • BMMSC isolation and culture Human BMMSC were isolated from 3-4ml of bone marrow aspirates using Ficoll density gradient. Cells were cultured in DMEM (1 g/L glucose) supplemented with 1% penicillin/streptomycin and 10% fetal bovine serum at 37C in 95% air with 5% carbon dioxide at 100% humidity. Medium was replenished every 3 days. Confluent cultures were passaged by trypsinization and gentle scraping. Cells at passage 5 were used in the experiments.
  • Human hepatocytes were cultured in DMEM (1g/L glucose) supplemented with 1 % non essential amino acids, 1% penicillin/streptomycin, 1 % glutamax and 10% fetal bovien serum at 37C in 95% air with 5% carbon dioxide at 100% humidity. Cells were cultured for a maximum of 15 days after which new cells were put in culture.
  • Human BMMSCs were seeded in 6-well format inserts at a density of 30000 cells. In the compartment underneath the inserts, human liver cells were seeded at the same density. Cells were replenished with fresh growth media every 3 days. The media used to replenish the cells was each cell growth media. Cells were harvested 10 days post co-culture. Human liver cells growth media: DMEM low glucose, non essential amino acids, fetal bovine serum, glutamax, penicillin/streptomycine.
  • BMMSC growth media DMEM low glucose, fetal bovine serum, peniciline/streptomycine.
  • Human liver cells were cultured in T75 flasks. After 48h of cell counfluency, the media was removed and filtered using 0.2um syringe filter and stored at -80C. The liver cells were then washed once after which a "removing reagent" was added to the cells ensuring total removal of cells while leaving the deposited ECM intact. The ECM was gently washed once with media (without FBS) to remove any traces of the removing agent and then the ECM was scraped in 2 ml of media (without FBS) and aliquoted and stored at -80C.
  • hepatocyte markers induced in BMMSC 1 - human hepatocyte cells express the various liver markers. 2- On the other hand, undifferentiated BMMSC do not express liver markers. 3- When BMMSC differentiate into hepatocytes these cells start expressing liver specific markers: AFP and p450.
  • BMMSCs takes place, which verifies that the alleged differentiation has taken place. Undifferentiated BMMSCs do not express p450 (1 ). After culturing BMMSCs on human liver cell ECM, p450 expression was induced (2) and increased induction was detected upon the treatment with 1 mM phenobarbital for 1 h (3) and 4h(4). House keeping gene was used to ensure equal loading of the samples.
  • FIG 13 shows the morphology of differentiated BMMSCs cultivated on human liver cell ECM.
  • BMMSCs cultured on plastic maintained their spindle shape morphology (right panel), where as when cultured on human liver ECM for 10 days these cells changed to round shaped cells (left panel).
  • Figure 14 the expression of Liver-specific connexins in differentiated stem cells (hepatocytes) can be seen. This indicates that the liver tissue is being generated.
  • Figure 15 confirms that the liver-differentiated BMMSC indeed work as hepatocytes with the specific induction of p450.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Rheumatology (AREA)
  • Hematology (AREA)
  • Immunology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

La présente invention se rapporte au domaine de la technologie des cellules souches. Cette invention concerne une méthode in vitro permettant l'induction de cellules d'origine humaine pour leur faire produire des matrices extracellulaires spécifiques de cellules et des facteurs de différenciation. Cet extrait est utilisable pour la différenciation des cellules souches humaines en cellules tissulaires désirées, comme par exemple la différenciation des cellules souches issues de moelle osseuse en chondrocytes.
PCT/EP2008/068110 2007-12-21 2008-12-19 Procédé de préparation de matrices extracellulaires spécifiques de cellules WO2009080794A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US1578607P 2007-12-21 2007-12-21
US61/015,786 2007-12-21
US10298608P 2008-10-06 2008-10-06
US61/102,986 2008-10-06

Publications (1)

Publication Number Publication Date
WO2009080794A1 true WO2009080794A1 (fr) 2009-07-02

Family

ID=40545824

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2008/068110 WO2009080794A1 (fr) 2007-12-21 2008-12-19 Procédé de préparation de matrices extracellulaires spécifiques de cellules

Country Status (1)

Country Link
WO (1) WO2009080794A1 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010146177A1 (fr) * 2009-06-18 2010-12-23 Mc2 Cell Aps Extrait extracellulaire de moelle osseuse et utilisation thérapeutique de celui-ci
EP2361971A1 (fr) * 2010-02-19 2011-08-31 Banc De Sang I Teixits Procédure pour obtenir de compositions formulées de tissu permettant la régénération dirigée de tissu osseux
CN108546675A (zh) * 2018-05-17 2018-09-18 广东芙金干细胞再生医学有限公司 促进骨髓干细胞分化为肝细胞的细胞外基质的制备方法
WO2018185260A1 (fr) * 2017-04-06 2018-10-11 Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Nouvelle matrice dérivée d'hépatocytes humains pour la différenciation de cellules souches et la réparation de tissus
US11274279B2 (en) 2020-03-11 2022-03-15 Bit Bio Limited Method of generating hepatic cells
CN114591900A (zh) * 2022-03-18 2022-06-07 广州捷创生物科技有限公司 一种提高干细胞分化能力的体外培养方法及培养基和应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040157323A1 (en) * 1996-12-10 2004-08-12 Badylak Stephen F. Biomaterial derived from vertebrate liver tissue
WO2007056547A2 (fr) * 2005-11-08 2007-05-18 Georgia Tech Research Corporation Materiau bio decellularise derivant de cellules souches d'embryons

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040157323A1 (en) * 1996-12-10 2004-08-12 Badylak Stephen F. Biomaterial derived from vertebrate liver tissue
WO2007056547A2 (fr) * 2005-11-08 2007-05-18 Georgia Tech Research Corporation Materiau bio decellularise derivant de cellules souches d'embryons

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
CHEN X ET AL: "HUMAN BONE MARROW STROMAL CELL CULTURES CONDITIONED BY TRAUMATIC BRAIN TISSUE EXTRACTS: GROWTH FACTOR PRODUCTION", JOURNAL OF NEUROSCIENCE RESEARCH, WILEY-LISS, US, vol. 69, no. 5, 2 August 2002 (2002-08-02), pages 687 - 691, XP009044236, ISSN: 0360-4012 *
EL-SABBAN MARWAN E ET AL: "Xenogenic bone matrix extracts induce osteoblastic differentiation of human bone marrow-derived mesenchymal stem cells.", REGENERATIVE MEDICINE JUL 2007, vol. 2, no. 4, July 2007 (2007-07-01), pages 383 - 390, XP002525452, ISSN: 1746-076X *
ENGLER ADAM J ET AL: "Matrix elasticity directs stem cell lineage specification", CELL, vol. 126, no. 4, August 2006 (2006-08-01), pages 677 - 689, XP002525451, ISSN: 0092-8674 *
LEE K-D ET AL: "In vitro hepatic differentiation of human mesenchymal stem cells", HEPATOLOGY, WILLIAMS AND WILKINS, BALTIMORE, MD, US, vol. 40, no. 6, 1 January 2004 (2004-01-01), pages 1275 - 1284, XP003018526, ISSN: 0270-9139 *
SANCHEZ-RAMOS J ET AL: "Adult bone marrow stromal cells differentiate into neural cells in vitro", EXPERIMENTAL NEUROLOGY, ACADEMIC PRESS, NEW YORK, NY, US, vol. 164, no. 2, 1 August 2000 (2000-08-01), pages 247 - 256, XP002199139, ISSN: 0014-4886 *
SCHWARTZ ROBERT E ET AL: "Defined conditions for development of functional hepatic cells from human embryonic stem cells", STEM CELLS AND DEVELOPMENT, ELSEVIER, AMSTERDAM, vol. 14, no. 6, 1 December 2005 (2005-12-01), pages 643 - 655, XP009090955, ISSN: 1547-3287 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010146177A1 (fr) * 2009-06-18 2010-12-23 Mc2 Cell Aps Extrait extracellulaire de moelle osseuse et utilisation thérapeutique de celui-ci
EP2361971A1 (fr) * 2010-02-19 2011-08-31 Banc De Sang I Teixits Procédure pour obtenir de compositions formulées de tissu permettant la régénération dirigée de tissu osseux
WO2018185260A1 (fr) * 2017-04-06 2018-10-11 Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Nouvelle matrice dérivée d'hépatocytes humains pour la différenciation de cellules souches et la réparation de tissus
CN108546675A (zh) * 2018-05-17 2018-09-18 广东芙金干细胞再生医学有限公司 促进骨髓干细胞分化为肝细胞的细胞外基质的制备方法
US11274279B2 (en) 2020-03-11 2022-03-15 Bit Bio Limited Method of generating hepatic cells
CN114591900A (zh) * 2022-03-18 2022-06-07 广州捷创生物科技有限公司 一种提高干细胞分化能力的体外培养方法及培养基和应用
CN114591900B (zh) * 2022-03-18 2022-10-18 河北北冥生物科技有限公司 一种提高干细胞分化能力的体外培养方法及培养基和应用

Similar Documents

Publication Publication Date Title
AU2003206844B2 (en) Pluripotent embryonic-like stem cells derived from teeth and uses thereof
Mondrinos et al. Engineering three-dimensional pulmonary tissue constructs
KR100907248B1 (ko) 분화된 어린 지방 세포와 생분해성 중합체의 이식에 의한신체의 부피 대체 방법
Harkness et al. Selective isolation and differentiation of a stromal population of human embryonic stem cells with osteogenic potential
US20090297579A1 (en) Control of Cells and Cell Multipotentiality in Three Dimensional Matrices
US20110207175A1 (en) Multi-culture bioreactor system
Mujaj et al. Serum-free primary human fibroblast and keratinocyte coculture
KR20040094910A (ko) 개선된 지방세포 분화된 지방 유래 성체 줄기세포 및 이의용도
JP2005521405A (ja) 単球を起源に持つ、脱分化したプログラム可能な幹細胞およびそれらの製造と使用
Danišovič et al. Comparative analysis of mesenchymal stromal cells from different tissue sources in respect to articular cartilage tissue engineering
US8084023B2 (en) Maintenance and propagation of mesenchymal stem cells
US20110052693A1 (en) Method for producing artificial skin
CN104263699A (zh) 细胞移植用临床治疗级真皮多能干细胞规模化制备培养方法
Farrell et al. A comparison of the osteogenic potential of adult rat mesenchymal stem cells cultured in 2-D and on 3-D collagen glycosaminoglycan scaffolds
RU2433172C2 (ru) Способ получения гомогенной популяции стволовых клеток и ее применение
Nair et al. Identification of p63+ keratinocyte progenitor cells in circulation and their matrix-directed differentiation to epithelial cells
WO2009080794A1 (fr) Procédé de préparation de matrices extracellulaires spécifiques de cellules
US9422522B2 (en) Method of producing adipocytes from fibroblast cells
New et al. Towards reconstruction of epithelialized cartilages from autologous adipose tissue‐derived stem cells
Dai et al. The human skin-derived precursors for regenerative medicine: current state, challenges, and perspectives
JP2022501118A (ja) 脂肪由来幹細胞およびゼラチンを含む生体材料ならびにその製造方法
WO2003050273A1 (fr) Milieu de culture pour des cellules humaines et methode de culture
Nazbar et al. Molecular imprinting as a simple way for the long-term maintenance of the stemness and proliferation potential of adipose-derived stem cells: an in vitro study
JP2005531298A (ja) 軟骨の欠損を修復するための再分化細胞
CN113166722A (zh) 羊水细胞来源的细胞外基质及其用途

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08865837

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08865837

Country of ref document: EP

Kind code of ref document: A1