NZ620290B2 - Methods and compositions for improving sperm functionality - Google Patents

Methods and compositions for improving sperm functionality Download PDF

Info

Publication number
NZ620290B2
NZ620290B2 NZ620290A NZ62029012A NZ620290B2 NZ 620290 B2 NZ620290 B2 NZ 620290B2 NZ 620290 A NZ620290 A NZ 620290A NZ 62029012 A NZ62029012 A NZ 62029012A NZ 620290 B2 NZ620290 B2 NZ 620290B2
Authority
NZ
New Zealand
Prior art keywords
sperm
conjugate
phosphatidyl
catalase
cholesterol
Prior art date
Application number
NZ620290A
Other versions
NZ620290A (en
Inventor
Bridget Lee Dicker
Jiwon Hong
Keith Hudson
Shakeela Nathalia Jayasinghe
Original Assignee
Androgenix Limited
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 Androgenix Limited filed Critical Androgenix Limited
Priority claimed from PCT/NZ2012/000140 external-priority patent/WO2013022354A1/en
Publication of NZ620290A publication Critical patent/NZ620290A/en
Publication of NZ620290B2 publication Critical patent/NZ620290B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • A01N1/0205Chemical aspects
    • A01N1/021Preservation or perfusion media, liquids, solids or gases used in the preservation of cells, tissue, organs or bodily fluids
    • A01N1/0221Freeze-process protecting agents, i.e. substances protecting cells from effects of the physical process, e.g. cryoprotectants, osmolarity regulators like oncotic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/06Tripeptides
    • A61K38/063Glutathione
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1774Immunoglobulin superfamily (e.g. CD2, CD4, CD8, ICAM molecules, B7 molecules, Fc-receptors, MHC-molecules)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/465Hydrolases (3) acting on ester bonds (3.1), e.g. lipases, ribonucleases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70596Molecules with a "CD"-designation not provided for elsewhere
    • 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
    • C12N5/0608Germ cells
    • C12N5/061Sperm cells, spermatogonia
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/96Stabilising an enzyme by forming an adduct or a composition; Forming enzyme conjugates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)

Abstract

The disclosure relates to a conjugate comprising a membrane anchoring agent, polyethylene glycol effective and a functional molecule that is in increasing the lifespan of sperm in the female reproductive tract, wherein the functional molecule is attached to the polyethylene glycol by an attachment group. Example conjugates are shown in the abstract figure. The functional molecule may be catalase, glutathione, CD55, CD59, CD73, DNaseI or SPAM1. The membrane anchoring agent may be a lipid, such as cholesterol, diacylglycerolipids, dialkylglycerolipids, glycerophospholipids, sphingosine derived diacyl- and dialkyl- lipids, ceramide, phosphatidate, phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl serine, phosphatidyl inositol or phosphatidyl glycerol. The attachment group may be ester amine reactive groups, maleimide, vinyl sulfone, iodoacetamide, orthopyridyl disulphide, hydrazide, benzotriazole, succinimidyl carbonate or succinimidyl active esters based on priopionic and butanoic acids. The disclosure also relates to a method for preparing a composition for use in artificial insemination or in vitro fertilization using sperm and the conjugate. roup. Example conjugates are shown in the abstract figure. The functional molecule may be catalase, glutathione, CD55, CD59, CD73, DNaseI or SPAM1. The membrane anchoring agent may be a lipid, such as cholesterol, diacylglycerolipids, dialkylglycerolipids, glycerophospholipids, sphingosine derived diacyl- and dialkyl- lipids, ceramide, phosphatidate, phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl serine, phosphatidyl inositol or phosphatidyl glycerol. The attachment group may be ester amine reactive groups, maleimide, vinyl sulfone, iodoacetamide, orthopyridyl disulphide, hydrazide, benzotriazole, succinimidyl carbonate or succinimidyl active esters based on priopionic and butanoic acids. The disclosure also relates to a method for preparing a composition for use in artificial insemination or in vitro fertilization using sperm and the conjugate.

Description

METHODS AND COMPOSITIONS FOR IMPROVING SPERM FUNCTIONALITY FIELD OF THE INVENTION This ation relates to methods for enhancing the functionality of sperm. More particularly, this application relates to methods for reducing the number of sperm required in livestock artificial insemination (Al) in particular for application with flow semen sexing. The methods may also be employed to increase the fertility of sperm in some human male individuals with sub—optimal fertility.
BACKGROUND The ability to identify and select male and female sperm has great value in the livestock industries, where there is an established market in Al of over US$12 billion annum in the Organization for Economic ation and Development (OECD) countries. This is particularly true in the dairy industry where over 80% of dairy farmers in key OECD markets impregnate their cows through Al. Sexed semen provides the opportunity to increase farmer productivity and income. For example, the availability of sexed semen has significant impact in reducing and/or eliminating the l returns of male dairy calves as compared to female calves. 2O Currently, the only commercial technique for semen sexing uses flow cytometry to sort sperm on the basis of DNA content. Bovine sperm bearing the Y chromosome have approximately 4% less DNA than sperm g the X chromosome. This difference, in combination with a fluorescent DNA binding dye (for example Hoechst 33342) and flow try, permits purification of X some bearing sperm to greater than 90% (Johnson et al., 1989). However, the ability to sort bovine sperm is currently limited to a rate of 8000 s‘1 which, when each straw, or close, contains 2 x 106 sperm, ates to 14 straws/hour (Sharpe and Evans, 2009). As a result, sexed semen straws generally incorporate ten-fold less sperm than unsexed straws. In on, the sorting process itself has a negative effect on the ity of the sperm. The reduction in the number of sperm per straw, together with reduction in sperm fertility due to the sorting process, causes a significant reduction of 14% in the conception rate for sexed sperm compared to unsexed sperm (Frijters et al., 2009). The sexed semen also has a significant price premium over d sperm due to the high cost of sorting even the sub-optimal number of sperm used in the sexed semen . A valuable addition to the semen sexing technology would be a method to e the fertility of sperm so that dose of considerably less than the approximately 2 x 106 sperm per straw currently used for sexed semen would achieve the same tion rate as the normal, unsexed, straws.
Such methods would also have application in swine AI where much higher doses of sperm are used in the standard AI methodology than with bovine, namely approximately 2500 x 106 sperm per straw. Recent work suggests that more sophisticated techniques involving deep intrauterine insemination can lower this requirement to 50-70 million (Vazquez et al., 2005; Vazquez et al., 2008). However, this reduced dose is still beyond the present commercial capability of flow cytometry sorted sperm.
The sperm y in the female reproductive tract Sperm are highly specialized cells that deliver the d male genome to the haploid female genome contained in the oocyte. Despite this seemingly simple mission, the path to achieving this goal is highly complex. Extraordinarily large numbers of sperm are inseminated in a l mating, for example approximately four billion sperm/oocyte in the cow. The inseminated sperm spend a variable period of time, ranging from hours to days in the different regions of the female reproductive tract (FRT). The environments that sperm encounter from ejaculation to fertilization of the oocyte also vary considerably.
These environments range from the complex molecular mix added to sperm at ejaculation by the male to the various female secretions and different cell es of the female epithelia (Drobnis and Overstreet, 1992).
Once sperm are deposited in the FRT, a combination of active sperm migration and female uterine muscle contraction propels the sperm to the oocyte. During the journey through the FRT, sperm can be retained in lized regions, most notably the cervix and t (Drobnis and Overstreet, 1992). This retention may increase the probability that at least some sperm will be present in the oviduct at the same time as ovulation occurs. However, for the cervix it is more likely that the restriction of entry and trapping acts as a negative ion imposed against sperm by the female. In fact, one of the major innovations that launched the modern bovine AI industry was the finding that considerably less sperm were required when sperm were passed through the cervix and deposited into the body of the uterus (Fcote, 2002). The final phase of the sperm y in the oviduct involves release of sperm from the isthmus region (controlled by the female) and travel to the ampulla for fertilization of the oocyte. At this time near unitary 2012/000140 numbers of sperm are present (Drobnis and Overstreet, 1992). Fertilization itself is again a complex phenomena involving penetration of the s oophorus and subsequently the zona pellucida (Kata et al., 1989). Although the sperm journey through the FRT is broadly similar between ian species, various aspects do differ.
Sperm also undergo a maturational change while resident in the FRT, known as capacitation. When sperm are ejaculated, they are not capable of fertilizing the oocyte.
However, during passage through the FRT sperm gain the ty to fertilize. Changes to sperm during passage through the FRT include alterations in membrane properties, enzyme ties and motility (Salicioni et al., 2007). One such ant change is the 1O loss of cholesterol from the outer sperm surface membrane (Flesch et al., 2001; Osheroff et al., 1999; ti et al., 1999). Ultimately these changes enable sperm to respond to stimuli that induce the acrosome reaction and penetration of the egg. One of the important changes that occur during capacitation is alterations in the e properties of sperm. A specialized protein-carbohydrate coating izes the surface membrane (Schroter et al., 1999), regulates tation r—Petersen et al., 1998), facilitates transport through the FRT (Tollner et al., 2008b), and enables attachment at the oviduct (Tollner eta1., 2008a). In ent species, essentially the same functions are canied out by the surface coatings, however the specific molecular components vary (Calvete and Sanz, 2007; Tollner et al., 2008a; Topfer-Petersen et al., 1998).
The attrition of sperm in the female reproductive tract In a natural bovine mating, approximately 4 billion sperm are inseminated yet less than 10,000 get to the oviduct and less than 10 get through to the oocyte (Mitchell et al., 1985). Why there are such large losses is poorly understood. Following coitis, greater than 80% of sperm are lost through vaginal discharge (Mitchell et al., 1985). The remaining sperm form a gradient in concentration from the cervix to the oviduct (Hawk, 1983; Hunter, 2003; Mitchell et al., 1985). In , approximately 10,000 sperm arrive at the t 6-8 hours after insemination (Mitchell et al., 1985). By 12 to 24 h after insemination, sperm have either been lost through back flushing, eliminated by phagocytosis or reached the oviduct (Hawk, 1983). In pigs, there is strong evidence for ytosis of sperm by polymorphonuclear neutrophils, with a massive infiltration of neutrophils occurring in the uterine lumen shortly after insemination (Matthijs et al., 2003). Recently, similar evidence that neutrophils infiltrate the uterine lumen after insemination in cows has been published (Alghamdi et al., 2009).
How sperm are d during passage through the female reproductive tract mental evidence suggests that both motile and damaged (immotile and/or dead) sperm are lost by discharge (Lightfoot and Restall, 1971; Oren—Benaroya et al., 2007). In contrast, in vitro evidence indicates that live sperm are preferentially phagocytosed by neutrophils (Woelders and Matthijs, 2001). Several phenomena contribute to sperm damage from the FRT, gh the mechanism and significance poorly understood. Such phenomena include: 0 Adhesive ties of female epithelia ing sperm and/or damaging the sperm surface, particularly mucus laden surfaces such as the cervix. This occurs by both biochemical and physical shearing (Katz et al., 1989; Mullins and Saacke, 1989). 0 Female secretions modulating and/or damaging the sperm surface or functionality such as flagella activity, tation and acrosome status. Such secretions include antibodies, complement components, molecular species affecting energy, c and oxidation homeostasis, ing molecules particularly for capacitation, and also lic entities. Microorganisms that are present in the FRT may also e agents that affect sperm.
Sperm also cause damage to themselves through generation of ve oxygen species (ROS) mainly as a by-product of mitochondrial function (de Lamirande and Gagnon, 1995; Koppers et al., 2008; Vernet et al., 2001). ROS cause loss of sperm motility and lipid peroxidation. The latter damage leads to alteration of the membrane properties such as flexibility and fluidity, and can also lead to lack of membrane integrity and/or decreased chromatin quality (Storey, 1997). Sperm are particularly sensitive to duced damage because of their membrane composition and their limited antioxidant defenses. In particular, the high proportion of saturated fatty acids (PUFA) in the surface membrane makes this membrane highly susceptible to oxidation (Jones et al., 1979). The nature of the sperm cell, with limited cytoplasmic fluid, also ains the availability of intracellular antioxidants (Koppers et al., 2008, & ref within). In human sperm at least, there exists a strong relationship between ROS production and antioxidant protection for determining the lifespan of sperm in the absence of external damaging agents (Alvarez and Storey, 1985; Storey, 1997, 2008).
Sperm motility, but not viability, is sensitive to specific ROS Sperm from human (Bell et al., 1992), bovine (Bilodeau et al., 2002), equine (Baumber et al., 2000) and porcine (Awda et al., 2009) all show loss of motility when exposed to H202 concentrations in the mico molar range, however where examined this loss of motility has not been associated with loss of viability (Awda et al., 2009; Bell et al., 1992). In addition this ROS-induced loss of motility is specific to H202 and not other ROS like 02' (Awda et al., 2009; Baumber et al., 2000; Bilodeau et al., 2002). The mechanism for H202—induced loss of ty is currently unknown, r it may be related to the observation that, unlike other ROS, H202 is able to pass through the cell membrane (Bienert et al., 2006). This membrane passage by H202 has so far been shown to be tated by aquaporin membrane proteins. Sperm express surface membrane aquaporin pumps, which are thought to be associated with sperm volume regulation by being able to pump H20 through the cell membrane (Chen and Duan, 2011; Chen et al., 2011; Young, 2010). ivity of sperm motility to H202 may also occur in the FRT. It has been shown that dead sperm in combination with aromatic amino acids produce H202 (Shannon and Curson, 1972; Tosic and Walton, 1950) and dead sperm are abundant in the FRT.
H202 may also result from bacterial organisms present in the FRT. In particular, Lactobacillus acidophilus is known to produce H202 and is tly present in both the human (Klebanoff and Smith, 1970) and the cattle vagina (Otero and Nader-Macias, 2006).
In all species where sperm suffer H202-induced loss of ty, antioxidants such as catalase and glutathione are able to rescue the loss of motility if added in sufficient tration and simultaneously with the H202 er et al., 2000; Bilodeau et al., 2002). In contrast, antioxidants reactive against 02‘ such as superoxide ase (SOD) could not rescue motility (Baumber et al., 2000; Bilodeau et al., 2002; Lapointe and Sirard, 1998).
Surface properties of sperm may have a significant influence on fertility The lized protein-carbohydrate coating that tates transport through the FRT (Tollner et al., 2008b), may influence fertility by protecting sperm from mucus capture in the FRT or assisting the motion of sperm through mucus. An example of a PCT/N22012/000140 protein that coats the surface of sperm and facilitates travel through the FRT is B-defensin 126 in macaque monkeys. This highly ated glycoprotein coats macaque sperm and is a major component of the sperm glycocalyx (Yudin et al., 2003; Yudin et al., 2005).
Importantly, this glycoprotein facilitates movement of sperm through cervical mucus (Tollner et al., 2008b) as does the human nsin 126 on human sperm through the al mucus surrogate, hyaluronic acid (Tollner et al., 2011). Men that are gous for a deletion mutation of Bidefensin 126 exhibit impaired sperm function and subfertility (Tollner et al., 2011). Macaque B-defensin 126 has extensive O-linked- glycosylation in the carboxy-terminal portion of the n and a significant amount of sialic acid on the carbohydrate-terminal residues (Yudin et al., 2005). The vely charged sialic acid residues from B-defensin 126 contribute the majority of the charge on the macaque sperm e (Yudin et al., 2005) and presumably also on human sperm er et al., 2011). These sperm surface charges may well be responsible for ation through the vely charged cervical mucus or substitutes with similar properties of charge and viscosity like hyaluronic acid (Aitken et al., 1992; Tang et al., 1999). If surface charge is important for sperm movement through mucus, changing either the actual surface charge or the distribution of charge may affect sperm motion in uterine mucus and fertility.
In summary, the FRT is hostile to sperm, in particular selecting for motile non- damaged sperm but also ng the vast majority of sperm. While in the FRT, sperm have to deal with a Wide variety of physiological environments, mature particularly at the cell surface and respond appropriately to signals at the right time and place. Thus despite the sperm’s simple mission and relatively simple construction, successful sperm have the characteristics of at least reaching the upper uterine horn, remaining undamaged (mainly surface phenomena), not being phagocytosed, remaining motile (a function of mitochondria, glycolytic enzymes and flagella components), avoiding capture by mucus and being able to respond to s appropriately (a surface phenomena but also involving signal transduction and effector pathways). Treatments to sperm that enhance the ability of sperm to remain undamaged, , not phagocytosed and fimctionally ent could therefore reduce the number of sperm required for insemination.
Pegylation 2012/000140 Polyethylene glycol (PEG) has the general a: H(OCH2CH2)nOH with typical molecular weights of SOD—20,000 daltons. It is non-immunogenic and soluble in aqueous ons. The polymer is nontoxic and generally does not harm active proteins or cells.
Pegylation of proteins has been shown to improve solubility and vascular longevity, and decrease the immunogenicity of xenogeneic proteins While retaining normal protein function (Abuchowski et al., 1977a; Abuchowski et al., 1977b; Jackson et al., 1987; Senior et al., 1991; Zalipsky et al., 1994). Pegylation has also been used directly on cells to provide immune camouflage, initially for transfusion of red blood cells 1D (Chen and Scott, 2001; Scott et 211., 1997) and subsequently for other tissues such as atic beta islet cells‘(Chen and Scott, 2001; ra and Iwata, 2009). For both red blood cells and pancreatic beta islet cells, the respective cell functions were preserved despite the pegylation.
Y OF INVENTION The present disclosure provides methods and conjugates for improving the functionality of cells, such as sperm. More cally, the present disclosure provides conjugates that can be employed to attach onal molecules of interest, such as proteins or carbohydrates, to cells. The disclosed methods and itions are effective in improving the functionality and/or fertility of sperm in the FRT, for e by preventing loss of motility, protecting t phagocytosis by neutrophils or other immune attack, facilitating sperm movement through the FRT by aiding movement or avoiding capture by mucus and thus in general extending the lifespan of sperm in the FRT and/or improving functionality. The disclosed methods and compositions can be employed in A1, for example, to reduce the number of sperm needed for insemination and to improve conception rates. Addition of proteins to cells other than sperm using the disclosed conjugates can also be used in other applications, such as lantation protection.
In one aspect, the t disclosure provides conjugates that can be employed to improve the onality of cells, such as sperm, by attaching a functional molecule of interest, such as a protein, to the surface of the cells. The disclosed conjugates comprise, or consist of, four components: a membrane anchoring component, such as a lipid; a spacer and/0r solubilizing component, such as PEG; an attachment group or linker; and a onal molecule of interest that is attached to the spacer and/or solubilizing component via the attachment group.
Lipids that can be effectively ed in the disclosed conjugates include cholesterol, diacylglycerolipids, dialkylglycerolipids, glycerophospholipids, sphingosine derived diacyl- and dialkyl- lipids, de, phosphatidate, phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl serine, phosphatidyl inositol and phosphatidyl glycerol. Examples of lipid-PEG—attachment group structures employed in the disclosed conjugates include those provided in Figs. lA—lC. In specific embodiments, the membrane anchoring component is cholesterol, the spacer and/or solubilizing component is PEG, the attachment group is an amine reactive group and the functional molecule of interest is catalase or glutathione.
In certain embodiments, functional molecules of st employed in the disclosed conjugates are able to increase the lifespan of sperm in the FRT by at least 20%, %, 40% or 50% compared to untreated sperm. Examples of such molecules include, but are not limited to, amino acids and their derivatives, hione, CD55, CD59, CD73, SPAMl, DNaseI, catalase, and variants thereof. The amino acid sequences of CD55, CD59, CD73, SPAMl, L3 and catalase from bovine are provided in SEQ ID NO: 1-6, respectively. Seminal plasma proteins that bind to the surface of sperm or other sperm surface proteins can also be used as the functional molecules of interest employed in the disclosed conjugates. In certain ments, the functional les of interest are polypeptides selected from the group consisting of: SEQ ID NO: 7~163 and variants thereof.
In another aspect, compositions comprising one or more of the conjugates disclosed herein and a physiologically acceptable carrier are also provided, together with preparations comprising at least one such composition and live sperm. In certain embodiments, the live sperm bear the X chromosome. Such ations can be effectively employed in Al or in vitro fertilization.
In a r aspect, methods for ing the functionality and/or fertility of sperm are provided, such methods sing contacting the sperm with an effective amount of a conjugate or composition disclosed . Such methods can be effectively ed with sperm from cows, pigs, sheep, goats, humans, camels, horses, deer, alpaca, dogs, cats, s and rodents. In certain embodiments, the sperm are sorted into X and Y chromosome-bearing sperm either prior to or after contact with the conjugate or composition.
In yet another aspect, the present disclosure provides methods for making a preparation for use in AI or in vitro fertilization, such methods comprising obtaining sperm from a mammal, ally separating the sperm into X chromosome-bearing and Y chromosome—bearing sperm, and contacting the sperm with an effective amount of composition and/or conjugate disclosed herein. Methods for separating X chromosome- bearing sperm from Y chromosome—bearing sperm are known to those of skill in the art, and include, for example, flow try. Such methods include, but are not d to, those described in US Patents No. 5,135,759, 5,985,216, 6,149,867 and 6,263,745.
Methods for the cryopreservation of sperm are also provided by the present disclosure. Such methods comprise: (a) contacting the sperm with a cryoprotectant and an effective amount of a composition and/or conjugate sed herein, and (b) storing the sperm and the composition/construct at a temperature of about 4°C to about -196°C, wherein the effective amount of the composition/conjugate is ent to increase the functionality and/or fertility of the sperm relative to sperm stored without the ition/conjugate. es of cryoprotectants that can be effectively employed in such methods include, but are not limited to, PEG, dimethyl sulfoxide , ethylene glycol, propylene glycol, polyvinyl idone (PVP), polyethylene oxide, raffinose, lactose, trehalose, melibiose, melezitose, mannotriose, stachyose, dextran, hydroxy—ethyl starch, sucrose, maltitol, ol and glycerol. In related s, preparations sing cryogenically preserved sperm and a composition and/or conjugate disclosed herein are ed. Methods for cryopreserving sperm are well known in the art and include those disclosed, for example, in US Patent 7,208,265 and US Patent Application Publication no.
US 2007/0092860.
The methods, compositions and constructs disclosed herein are particularly advantageous in the preparation of semen for use in A1 of mammals including, but not limited to, cows, pigs, sheep, goats, humans, camels, horses, deer, alpaca, dogs, cats, rabbits and rodents. Semen used in such methods may be either fresh ejaculate or have been usly frozen and subsequently thawed.
These and additional features of the present invention and the manner of obtaining them will become apparent, and the invention will be best tood, by reference to the following more detailed description.
BRIEF DESCRIPTION OF THE DRAWINGS Figs. lA—C show the structure of a tripartite le disclosed herein, with Fig. 1A showing the l structure, Fig. 1B showing the structure where X = NHS (ester amine reactive group) and Fig. 1C showing the structure where X = ester amine reactive group plus cein.
Figs. 2A-D illustrate binding of a cholesterol—PEGSOOO-FITC-catalase conjugate to sperm as determined by flow cytometry, with Fig. 2A showing flow cytometry analysis of freshly washed sperm, Fig. 2B showing flow cytometry analysis of sperm d with 1O catalase but without the cholesterol-PEGSOOO-FITC linker, Fig. 2C showing flow cytometry analysis of sperm treated with cholesterol-PEGSGOO-FITC-catalase, and Fig. 2D showing flow cytometry analysis of sperm treated with PBS alone. s shown are live cells that were negative for Hoechst 33258.
Figs. 3A-D illustrate binding of a cholesterol-PEGSCOO-FITC-catalase conjugate to Jurkat cells as determined by flow cytometry, with Fig. 3A showing flow cytometry is of untreated Jurkat cells, Fig. 3B showing flow cytometry is of Jurkat cells with catalase but without the cholesterol-PEGSOOO-FITC linker, Fig. 3C showing flow cytometry analysis of Jurkat cells treated with PBS alone, and Fig. 3D showing flow cytometry analysis of Jurkat cells with terol-PEGSOOO-FITC-catalase. Results shown are live cells that were negative for Hoechst 33258.
As outlined above, the present disclosure provides methods for improving the fimctionality and/or fertility of sperm, together with compositions and ates for use in such methods. In n embodiments, the methods and compositions disclosed herein enhance sperm motility, protect sperm from ytosis, aid sperm in avoiding capture by mucus, extend the lifespan of sperm in the FRT, and/or enhance the fimction of a ary sperm characteristic. The ability of a composition or conjugate to se the fimctionality and/or fertility of sperm may be determined by contacting sperm with the composition or conjugate; measuring parameters such as the motility, resistance to neutrophil attack, membrane integrity and/or presence of sperm surface markers indicative of capacitation and acrosome status on the treated sperm and ability to r from cryopreservation; and comparing the results with those obtained for untreated sperm. The functionality of sperm can also be determined by investigating their interaction with cervical mucus/explants or synthetic ues, and/or their y to capacitate, acrosome react and ize in vitro. Techniques for measuring the above parameters are well known in the art and include those described below. In certain embodiments, the disclosed methods comprise ting the compositions and/or conjugates disclosed herein with either sorted or unsorted sperm.
Conjugates The present disclosure provides methods for adding a functional molecule of interest to the surface of cells, such as sperm, using a conjugate including a membrane ing agent, PEG and the le of st. Such conjugates provide protection or enhancement of sperm functionality while at the same time allowing sperm to maintain the array of molecular and cellular ctions that occur in ascent through the FRT. In n embodiments, the disclosed conjugates have the following l structure: membrane anchoring agent—PEG—X-functional molecule, where X is any reactive group (referred to herein as an attachment group) that allows conjugation of at least one functional molecule of st.
As used herein, the term “membrane anchoring agent” or “membrane anchoring component” refers to a molecule that is known to spontaneously and stably incorporate into lipid bilayers, including cell membranes. Examples of such molecules include, but 2O are not limited to, the synthetic molecules described in US Patent Publication no. US 2007/0197466, the disclosure of which is hereby incorporated by reference. In n embodiments, the ne anchoring agent is a lipid. Lipids that may be effectively ed in the disclosed methods include, but are not limited to, diacyl- and dialkyl— glycerolipids, including glycerophospholipids and sphingosine derived diacyl- and dialkyl lipids, including ceramide. In n embodiments, the lipid is selected from the group consisting of: cholesterol, diacylglycerolipids, phosphatidate, phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl serine, phosphatidyl inositol and phosphatidyl glycerol. The lipid may be derived from one or more cis-desaturated fatty acids.
Cholesterol is considered to be a desirable lipid membrane anchoring agent as this lipid is the most abundant molecule in sperm surface membranes (Flesch et al., 2001) and is lost upon capacitation in the oviduct. Thus the same mechanism that removes endogenous cholesterol (cholesterol chelating agents such as bovine serum albumin; BSA) may also remove some ofthe added conjugate before fertilization.
PEGs having a wide range of lengths, or molecular weights, and a varying number of es can be effectively employed in the disclosed conjugates. For example, in certain embodiments the PEG has a molecular weight in the range of about 200 to about 40,000 daltons. PEGs contemplated for use in the conjugates disclosed herein include, but are not limited to, PEGs having one or more amine reactive groups that allow conjugation to a protein, and include linear and branched chain PEGs. As will be appreciated by those of skill in the art, when a PEG having an amine reactive group is employed in the conjugate, the ment group (X) is the amine reactive group.
In certain embodiments, the attachment group (X) is an amine ve group, however the attachment group can be any group that reacts with —COOH, -OH and/or -SH groups as well as disulfide (-S-S-) bonds and oxidized ydrates, on proteins or small molecules (Greenwald et al., 2003; s et al., 2002). Examples of reactive groups that have usly been used to attach PEG to proteins or peptides are shown in Table 1 below. Alternatively, the ne anchoring agent-PEG backbone can be linked to the functional molecule using a biotin-streptavidin linkage or click chemistry (Lutz and Zarafshani, 2008).
Table 1: Examples of reactive groups used to attach PEG to specific groups on proteins Reactive group previously used to attach PEG to proteins Protein reactive target (see Jevsevar et al., 2010; s et al., 2002 and references within) Thiol gSH—R) maleimide Thiol SH-R) vin_yl sulfone Thiol SH—R iodoacetamide Oxidized carboh drate residue hydrazide Histidine residue benzotriazole Histidine e succinimid 1- carbonate Amine group (HzN-R) succinimidyl active esters based on to ionic and butanoic acids An example of the general structure of a cholesterol-PEG-attachment group starting tripartite molecule is shown in Fig. 1A, where X = any attachment group.
Examples of specific starting tripartite les wherein X = NHS (ester amine reactive group) and X = ester amine reactive group plus fluorescein are shown in Figs. 1B and 1C, respectively. Tripartite terol-PEG—X molecules are available commercially and include, for example, those available from Nanocs Inc. The tripartite. molecule is initially covalently attached via the attachment group to the functional molecule of st, such as catalase. After attachment of the fimctional le of interest (cg. catalase) and purification, the conjugate can be contacted with cells, such as sperm, whereby the conjugate binds to the surface of the sperm.
Functional molecules of interest that can be added to sperm using the disclosed conjugates include amino acids and their derivatives, polyamino acids, peptides, s, adhesion molecules, immune proteins and antigens. Specific examples of functional 1O molecules of interest include antioxidants such as se and glutathione. Catalase and glutathione both protect sperm from H202, and if sperm are exposed to H202 in the FRT, membrane attached oxidation protection would aid sperm motility.
Other examples of onal molecules that can be effectively ed in the disclosed methods and conjugates include les that potentially t sperm from immune attack, ing CD55 (decay factor), CD59 and CD73 (Kirchhoff and Hale, 1996); or entrapment by neutrophils, such as DNasel (Alghamdi and Foster, 2005).
Membrane bound CD55 and CD59 inhibit the formation of complement induced membrane attack complex and could protect cells or sperm from ment attack (Fraser et al., 2003). CD55 and CD59 are glycosylphosphatidylinositol (GPI) linked proteins present on the surface of sperm and have specific roles in sperm function (Donev et al., 2008). Addition of proteins such as CD55 and CD59 to the cell surface also has application in other uses, such as transplantation protection (Hill et al., 2006). DNasel present in bovine seminal fluid is known to be associated with sperm fertility (Bellin et al., 1998; McCauley et al., 1999).
Other molecules that are important for fertility and that can also be employed in the sed methods include SPAMl, which is also GPI linked and present in the epididymis (Kirchhoff et al., 1997). SPAMl is a potential sperm adhesion molecule and hyaluronidase that enables sperm to penetrate through the hyalurom'c acid-rich cumulus cell layer nding the oocyte (Lathrop et al., 1990).
If sperm surface charge is important for movement of sperm h the FRT then altering surface charge or charge distribution could improve fertility. Using the conjugate» bed here and a functional group composed of amino acids, amino acid derivatives, polymeric amino acids or peptides enables sperm surface charge to be manipulated. For example, reacting an amine ve cholesterol-PEG with glycine would allow addition of one negative charge per conjugate, reaction with glutamic acid adds two negative charges per conjugate, y—carboxy~glutamic acid adds three ve charges per conjugate, and poly(L-glutamic acid) in defined numbers of residues, such as 20 or 50 able from Almanda rs), allows addition of 21 or 51 negative charges per conjugate, tively.
In general, apart from sperm charge modification, the seminal fluid (Novak et al., 2010a; Novak et al., 2010b; Rodriguez—Martinez et al., 2011) or epididymal proteins (Belleannee et al., 2011) that bind to sperm, or proteins present on sperm, and are ’10 correlated with high fertility (D'Amours et al., 2010; Novak et al., 2010b) represent functional molecules that can be employed in the disclosed methods and conjugates for addition to sperm.
The addition of GPI lipid anchored proteins to sperm during sperm maturation occurs at least partly through a mechanism where epididymosomes transfer such proteins to the sperm surface (Frenette et al., 2006; Kirchhoff and Hale, 1996). Epididymosomes are small membranous vesicles secreted by epithelial cells within the luminal compartment of the epididymis (Girouard et al., 2009). The inventors believe that the addition of a cholesterol-PEG-functional group construct to sperm is analogous to the transfer of such nked proteins that occurs in the epididymis. ptides and Proteins Proteins and/or polypeptides employed in the disclosed s, compositions and conjugates can be ed from l fluid (Kelly et al., 2006; Novak et al., 2010a; Novak et al., 2010b), epididymis or accessory sex glands (Moura et al., 2006a, 2007; Moura et al., 2006b) or other sources (eg. catalase from bovine liver (Sumner and Dounce, 1937)), or are commercially available. Alternatively, such proteins and/or polypeptides can be prepared recombinantly by inserting a polynucleotide that encodes the protein into an expression vector and expressing the antigen in an appropriate host.
Any of a variety of expression vectors known to those of ordinary skill in the art may be employed. sion may be ed in any appropriate host cell that has been transformed or transfected with an expression vector containing a DNA molecule that encodes a recombinant polypeptide. Suitable host cells include prokaryotes, yeast and WO 22354 higher eukaryotic cells. Preferably, the host cells employed are E. coli, mycobacteria, insect, yeast or a ian cell line such as COS or CHO.
The proteins and/or polypeptides employed in the methods, compositions and conjugates disclosed herein are isolated and purified, as those terms are commonly used in the art. Preferably, the proteins and/or polypeptides are isolated to a purity of at least 80% by weight, more preferably to a purity of at least 95% by weight, and most preferably to a purity of at least 99% by weight. In general, such purification may be achieved using, for example, the rd techniques of ammonium sulfate fractionation, SDS-PAGE electrophoresis, and affinity chromatography. 1O The conjugates and compositions disclosed herein encompass variant ptide sequences that have been d by one or more amino acid deletions, additions and/or substitutions. Variant sequences preferably exhibit at least 75%, more preferably at least 80%, more preferably at least 85%, more ably at least 90%, more preferably yet at least 95%, and most preferably at least 98% identity to a specific ptide sequence disclosed herein. The percentage identity is determined by aligning the two sequences to be compared as described below, determining the number of identical residues in the aligned portion, ng that number by the total number of residues in the inventive (queried) sequence, and multiplying the result by 100. In addition to exhibiting the recited level of sequence identity, t sequences preferably exhibit a functionality that is ntially similar to the functionality of the specific sequences disclosed herein.
Preferably a variant polypeptide sequence will have at least 80%, more preferably at least 85%, more preferably at least 90%, more preferably yet at least 95%, and most preferably 100% of the sperm fertility enhancing activity possessed by the specifically fied polypeptide sequence in one or more sperm fertility assays, such those described below.
Such variants may generally be identified by ing one of the polypeptide sequences disclosed herein, and evaluating the properties of the d polypeptide using, for example, the representative procedures described herein.
In certain embodiments, variant sequences differ from the specifically identified sequence only by conservative substitutions, deletions or modifications. As used , a "conservative substitution" is one in which an amino acid is substituted for another amino acid that has similar properties, such that one skilled in the art of peptide chemistry would expect the secondary structure and athic nature of the polypeptide to be substantially unchanged. In general, the following groups of amino acids represent conservative changes: (1) ala, pro, gly, glu, asp, gin, asn, ser, thr; (2) cys, ser, tyr, thr; (3) val, ile, leu, met, ala, phe; (4) lys, arg, his; and (5) phe, tyr, trp, his. Variants may also, or alternatively, contain other modifications, ing the deletion or addition of amino acids that have minimal influence on the nic properties, secondary structure and hydropathic nature of the polypeptide. For example, a polypeptide may be conjugated to a signal (or ) sequence at the N-terminal end of the protein which co- translationally or post-translationally directs transfer of the protein. The polypeptide may also be conjugated to a linker or other sequence for ease of synthesis, purification identification of the polypeptide (e.g., poly-His), or to enhance binding of the polypeptide 1O in the conjugate.
Polypeptide sequences may be aligned, and percentages of identical amino acids in a ed region may be determined against another polypeptide, using computer thms that are publicly available, such as the BLASTP algorithm. BLASTX and FASTX algorithms compare nucleotide query sequences translated in all reading frames against polypeptide sequences. The use of the BLAST family of algorithms is described at NCBI’s website and in the ations of Altschul et a1. (Altschul et a1., 1990; Altschul et al., 1997). The “hits” to one or more database sequences by a queried sequence produced by , BLASTP, FASTA, or a similar algorithm, align and identify r portions of sequences. The hits are ed in order of the degree of similarity and the length of sequence overlap. Hits to a database sequence generally represent an overlap over only a fraction of the sequence length of the queried sequence.
Methods In certain embodiments of the disclosed methods, sperm are purified by a single density layer (PercollTM PLUS, GE care, see protocol below). Sperm are then incubated in a suitable media with an effective amount of one or more of the compositions and/or conjugates disclosed herein for a short period of time, followed optionally by the addition of a le extender to enable immediate use or freezing.
Alternatively, the compositions and/or conjugates are added directly to the ate and, after slight dilution, a short incubation (15-30 minutes) and the addition of extender, resulting e is either cooled or frozen for e. In another method, the compositions and/or conjugates are added to extended semen. In other embodiments, sperm are sexed by flow cytometry and are collected in media containing an effective amount of one or more of the disclosed compositions and/or conjugates. Alternatively, once sufficient sorted sperm are collected, the composition and/or conjugate is added and the ing mixture is incubated in a suitable media for a short period of time, followed by the addition of extender and then either immediate use or freezing.
As used , the term “effective amount” of a composition and/or conjugates disclosed herein refers to that amount sufficient to enhance sperm motility, protect sperm from phagocytosis, allow sperm to avoid capture by mucus, extend the lifespan of sperm in the FRT, and/or se sperm functionality by at least 5-50% ed to untreated sperm.
Those of skill in the art will appreciate that for use in the disclosed methods, the compositions and conjugates disclosed herein may be present in compositions including one or more physiologically acceptable carriers or diluents, such as water or saline. Such compositions may additionally contain other components, such as preservatives, stabilizers, buffers and the like. Carriers, diluents and other components suitable for use in the present compositions are well known to those of skill in the art and include those currently used in preparations for AI.
All U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, n patent applications, non~patent publications, tables, sequences, web pages, or the like referred to in this specification, are incorporated herein by nce, in their entirety. The following examples are intended to illustrate, but not limit, this disclosure.
Example 1 Preparation and analysis of cholesteroLPEG-catalase Equal volumes of 2 mM terol—PEGSOOO—NHS—FITC (Nanocs) and 20 uM bovine catalase (Sigma; 100:1 ratio of cholesterol—PEGSOOO-NHS-FITC to bovine catalase) in phosphate buffered saline (PBS) were mixed by rotation for 3 hr at room ature. The mixed solution was then dialysed into PBS using a SOkDa molecular weight cut off (MWCO) membrane at 4°C overnight. The free cholesterol-PEGSOOO- NHS-FITC that had not reacted with the catalase was removed using an ammonium sulphate precipitation where 200 pl of 4.1 M ted ammonium te solution was added slowly into 500 pl of the cholesterol-PEGSOOO-NHS-FITC/catalase mixture.
PCT/NZZOIZ/000140 Centrifugation of the sample mixture for 20 min at 20,000 X g separated the free cholesterol-PEGSOOO-NHS-FITC as a pellet, and the reacted cholesterol-P1365000-FITC- catalase as atant. Both supernatant and pellet were then ed into PBS using a 10kDa MWCO membrane at 4°C overnight. Dialysed samples were analysed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) under reducing conditions. The resulting gel was ted to fluorescence imaging using an ImageQuant LAS—4000 (GE Healthcare), and stained with the Coomassie Brilliant Blue R—250 to visualize the protein bands. This ed gel analysis indicated that the catalase had been labeled with terol-PEG and that the cholesterol-PEG that had not reacted with se was removed by precipitation (cholesterol-PEG-catalase >95% purity). The e number of terol-PEG molecules added per catalase monomer was determined by measuring the fluorescence of known protein amount of cholesterol- PEGSOOO-NHS-FITC and employing a standard curve of free cholesterol-PEGSOOO~ NHS-FITC concentration versus fluorescence. Depending upon the ation of cholesterol-PEG-catalase, the range varied from 4.0 to 5.4 molecules of cholesterol-PEG per catalase monomer or 4 X this number for the intact tetramer.
Recovery of the cholesterol-PEGS000—FITC—catalase was determined by Coomassie staining and catalase activity assay. For the catalase activity assay, 0.5 ul of test s and serially diluted bovine catalase (Sigma) were placed in a microtitre plate, and incubated with 50 ul of 10 ng/ml of streptavidin-HRP (Biosource) and 50 ul of TMB substrate (Invitrogen) for 5 min. The plate was read at 450 nm on an EnVision plate reader (PerkinElmer) after stopping the reaction with 50 ul of 2 M sulfuric acid. Catalase activity of the test sample was calculated from the standard curve generated with known concentrations of bovine catalase. The catalase activity assay showed 85% recovery (equivalent to 1.11 mg; 44,406 units) of catalase in the form of cholesterol-PEGSOOO- FITC—catalase.
Example 2 Binding of cholesterol-PEG-catalase to sperm 1.5 ml of bovine sperm in liquid extender was carefully placed on top of 4 ml of 60%_PercollTM PLUS (GE Healthcare) , and centrifugated for 20 min at 700 x g at °C. The resulting purified sperm pellet was resuspended in non-capacitating media (NCM; see Table 2) containing 0.1 mg/ml of BSA to a cell concentration of 5 X 107/ml. 200 pl of the d sperm was mixed with an equal volume of 3 mg/ml cholesterol- PEGSOOO—FlTC-catalase, and incubated light protected at 37°C for 30 min. The unbound cholesterol-PEGSGOO-FITC-catalase was removed by layering 400 pl of sperm/cholesterol-PEGSGOO-FITC—catalase mixture on the top of 500 pl of 60% PercollTM PLUS , followed by centrifugation for 20 min at 700 X g at 20°C. The sperm pellet was resuspended in NCM containing 0.1 mg/ml of BSA to a total volume of 400 1.11.
The g of cholesterol-PEGSOOO-FITC-catalase to sperm was analysed by flow cytometry and catalase activity assay. For flow try analysis, the cholesterol- PEGSOOO-FITC-catalase bound sperm were diluted to 5 X 106 cells/ml in NCM containing 0.1 mg/ml of BSA, and stained with 0.2 gig/ml of a viability dye (Hoechst 33258). Significant binding (approx. 10 fold over background) of terol-PEGSOOO» FlTC-catalase to live sperm (Hoechst 33258 negative) was observed (see Fig. 2C). The catalase activity of cholesterol—PEGSOOO-FITC-catalase bound sperm was measured as bed in Example 1. 79 units of catalase activity were measured in 2.5 X 106 sperm cells after binding with cholesterol—PEGSGOO-FITC—catalase, s no catalase activity was detected in control sperm without addition of cholesterol-PEG5000-FITC-catalase.
Table 2: 1X NCM (non-capacitating media, pH 7.4) Component Concentration MgClz Sodium o ruvate HEPES NaCl Lactate_@5%) Gentamicin Example 3 Binding of cholesterol-PEG-catalase to Jurkat cells Jurkat cells (ATCC, 2; human T lymphocyte cell line) were cultured in RPM-1640 media with 10% fetal calf serum. For analysis, cells were removed from culture, centrifuged at 700 X g for 5 min, the culture media was removed and cells were resuspended at a concentration of 5 X 107 cells/ml in PBS. 100 pl of the Jurkat cell sion was mixed with 100 ul of 2 mg/ml cholesterol-PEGSOOO-FITC-catalase and incubated light protected at 37°C for 30 min. Cells were centrifuged for 5 min at 700 X g, WO 22354 supernatant removed, and resuspended in 400 pl of fresh PBS. The Jurkat cells were analyzed by both flow cytometry and catalase activity assay.
For flow cytometry analysis, the Jurkat cells were d in 200 pl of PBS to a concentration of 5 X 106 cells/mi. 0.2 gig/ml of Hoechst 33258 (Invitrogen, H21491) was then added to each sample (see Fig 3).
The catalase activity of the treated Jurkat cells was measured as described in Example 1. 50 pl of 5 X 107 cells/m1 of each test sample was analysed in the assay and the catalase activity contained in each of the Jurkat samples was calculated from the standard curve. The catalase activity assay indicated: 0 U of catalase in freshly washed 1O Jurkat cells, 44 U in Jurkat cells with se alone without linker, 66 U in Jurkat cells with choiesteroi-PEGSOOO-FITC—catalase (units per 2.5 X 106 cells).
Example 4 Cholesterol-PEG-catalase protects sperm from H202 induced loss of motility In this experimental configuration, a tration of H202 is chosen that causes sperm to rapidly lose motility (30-60 minutes) unless oxidation protection like catalase is present. Sperm that have had cholesterol-PEG—catalase added and been washed so that no remaining free cholesterol-PEG—catalase remains are compared with sperm that have been exposed to a similar molar amount of catalase as contained in the cholesterol-PEG- catalase and then , and also with sperm exposed to no catalase, for their y to resist H202 induced ty loss. Sperm motility is ined by a quantification system such as QualiSpermTM (Biophos).
Example 5 Identification of seminal plasma proteins in bovine seminal plasma a) Preparation of bovine seminal plasma Bovine semen samples were collected from three bulls using an artificial vagina and pooled. The pooled semen was centrifugated for 15 min at 1500 X g to remove sperm cells and the resulting supernatant was further centrifugated for 15 min at 15000 X g to remove any particulates. Complete mini protease tor cocktail was added to the cleared seminal plasma before storage at -20°C. Protein concentration was measured by honinic acid (BCA) protein assay kit (Pierce). b) Peptide preparation PCT/N22012/000140 Two s were used to prepare peptides from bovine seminal plasma for proteomic is. The first method used standard in-solution digestion of proteins.
Briefly, the seminal plasma was diluted in lysis buffer consisting of 7 M urea, 2 M thiourea, 4% CHAPS and 2 mM DTT, and incubated for 1 hr at 4°C with constant rotation. Following centrifugation at 14000 X g for 5 min at 4°C, an aliquot was removed for protein estimation by EZQTM protein quantitation kit (Molecular Probes). 30 pl of d seminal plasma containing approximately 100 pg eins was alkylated for 30 min with 2-fold molar excess of iodoacetarnide relative to the DTT. Proteins in the seminal plasma were then precipitated with methanol/chloroform. The resulting protein pellet was reconstituted in 0.5 M TEAB and l M urea containing 0.1 mg/ml trypsin, and incubated overnight at 37“C. Filter-aided sample preparation method (FASP II) ewski et al., 2009) was the second method used for peptide preparation from seminal plasma.
Acetonitrile was added to 5% to all peptide samples, before acidifying the peptides to pH 2 to 3 with formic acid. Peptides were then desalted on Sep-Pak Vac tC18 solid phase extraction cartridge (Waters), and completely dried in vacuum concentrator. c) Proteome analysis Dried peptide s were sent to the Australian me analysis facility (APAF), provided by the Australian Government through the National orative Research Infrastructure gy (NCRIS). At APAF, high sensitivity amino acid analysis was d out to accurately measure the amount of peptides in each sample. Peptide samples were loaded onto a Capillary LC system coupled to an MS/MS ment. The peptides were separated using a reverse phase C18 column and directly eluted into a Q- STAR mass spectrometer. lD-LC-ESI-MS data acquired was analysed by ProteinPilot software 3.0 (AB1) to identify the proteins. A thorough identification search was conducted in ProteinPilot. The International Protein Index (IPI) Bos taurus database (v3.49) was used for all searches. ns identified from two samples prepared by two different peptide preparation methods were ed to each other. Ensembl genome browser (www.cnsembl.org) was used to check the presence of transmembrane domains and signal peptide sequence in each identified protein.
A list of 73 seminal plasma proteins that may enhance functionality and/or fertility of sperm is provided in Table 3. The sequences for these proteins are provided in SEQ ID NO: 7-79, respectively. These 73 proteins were detected in two independently prepared seminal plasma samples, and also only those predicted to have signal peptide sequences were selected. Three seminal plasma proteins that were detected and that had multiple transmembrane domains were excluded from the list.
Table 3: Seminal plasma proteins detected by mass spectrometry and that also have signal ces n description WIidentifier Ensembl n ID MSLN MSLN protein IPIOO696375 P00000000202 GUSB GUSB protein ENSBTAP00000000941 STSGALI Sialylu‘ansferase 4A @91968 9100692648 ENSBTAP00000001538 LTF Lactotransferrin IPIOO710664 ENSBTAP00000001704 LGALS3BP Galectin-3—binding n IPIOO715562 P00000001802 PLODI Procoilagen—lysine,2-oxoglutarate 5-dioxygenase 1 IPIOO718774 GLIPRlLl GLIPRl-like protein 1 IPIOO713992 ENSBTAP00000006002 SPPl Osteopontin IP100691887 ENSBTAP00000006923 MANZBZ similar to idase, alpha, class 2B, member IP100709348 ENSBTAP00000008532 NGF Beta-nerve growth factor TPIOO685556 ENSBTAPOOOOOOO9796 fiComplement factor B (Fragment) WP—I007l7527 POOOOOOO9800 SPADHZ spermadhesin 2 IP100696725 ENSBTAPOOOOOO 10565 ENPEP Glutamyl aminopeptidase —i IPIOO685116 4 ENSBTAP00000010972 L§ETPINE2 Serpin peptidase inhibitor, clade E (Nexin, Max:839 ENSBTAP00000011485 plasminogen activator inhibitor type 1), member 2 IfEEGl Cellular repressor ofBIA-stimulated genes J j 1 IPIOO702458 ENSBTAPOOOOOOI 1757 ST6GAL1 Beta—galactoside alpha-2,6esialyltransferase IP100692543 ENSBTAP00000012565~l rain C—C motifchemokine 2 IPIOO690357 r ENSBTAPOOOOOOBE SPADHI Spermadhesin—l IPIOO688936 BNSBTAP00000014297 I‘IMPZ oproteinase inhibitor 2 ENSBTAP00000014476 ASAHl Acid ceramidase 1P100685320 W__IP100698673 OGN Mimecan 1P1007 16123 132M Beta~2-microglobulin 19100686769 STCH Heat shock 70 kDa protein 13 85695 CRISP3 Cysteine-rich secretory protein 3 IP100715999 ENSBTAP00000017l67 WFDCZ WAP four-disulfide core domain 2 IP100702630 ENSBTAP00000018498 CST6 .Cystatin E/M IL19100705340 ENSBTAP00000019583 PTGDS Prostaglandin—HZ D-isomerase IPIOO709683 ENSBTAP00000020065 L\7NN1 heinase 97935 ENSBTAP00000020086 m1 "‘TPIOO721428 ENSBTAPOOOOOO20469 C15H1 10RF34 Placenta-expressed transcript 1 protein IP100696232 ENSBTAPOOOOOO20999 TFPIZ Tissue factor pathway inhibitor 2 IP100709321 ENSBTAP00000021062 SCGBZAZ Z n IPIOO711254 ENSBTAP00000021195 GAA Lysosomai alpha-glucosidase IP100695601 ENSBTAPOWOOOZISZS ARSA Arylsulfatase A 19100713745 ENSBTAP00000021364 VNN2 vanin 2 IP100698407 TEXIOI TEXlOl protein IPIOO694179 PPIB Peptidyl-prolyl cis-trans isomerase B 02098 CTSL2 Cathepsin Ll IP100687440 933mm CTSS Cathepsin s IPIOO702008 C3 Complement C3 (Fragment) IP100713505 ENSBTAP00000022979 NUCB2 Nucleobindin 2 IP100696729 ENSBTAP00000023221 Wmdeoxyribonuclease I—like 3 jIPIOO709234IP100696577 ENSBTAP00000024347 BSPHI Seminal plasma protein BSP~30 kDa ENSBTAP00000025134 PLBDZ Putative phospholipase B-Iike 2 IP100702401 POOOOOO25343 PLA2G7 Platelet-activating factor hydroiase @0699458 ENSBTAP00000025719 ANG Arigiogenin-l IPIOO7 10136 ENSBTAP00000026126 PIGR Isofoml Long ofPolymeric immunoglobulin receptor IPIOO696714 GSN Gelsolin [PIOO694255 ENSBTAP00000026534 ENPP3 Ectonucleotide pyrophosphatase/phosphodiesterase 1131007 12650 ENSBTAP00000026900 family member 3 NPCZ Epididymal secretory protein E1 IPIOO71 1862 P00000029271 N'PNT r to Nephronectin precursor IP100826312 ENSBTAP00000029938 FAM3C FAM3C protein IPI00714868 ENSBTAP00000030039 947 Seron'ansferrm-like IPIOO705493 ENSBTAP0000003 l 846 RNASEI Seminal ribonuclease 19100700712 ENSBTAP00000036091 AZGPI Zinc-alpha—Z—glycoprotein IP100698993 i ENSBTAPOOOOOO37042 CTSA Lysosomal protective protein IP100687092 S moms protein IPIOO692789 NPPC C-type natriuretic peptide GPXS glutathione peroxidase s IPIOOS40765 SCGBIDZ Secretoglobin, family ID, member 2 IP100824879 i—ficiGALu UDP-GalzbetaGlcNAc beta 1,4- IPIOO690138 ENSBTAP00000044479 galactosyltransferase, polypeptide 4 ACRBP similar to sp32 $007168” ENSBTAP00000045557 CDHI CDHl protein IPIOO711327 P00000048482 PEBP4 Phosphatidylethanolamine-binding n 4 ENSBTAP00000050912 ACE 150 kDa protein IP100923883 ENSBTAP00000053314 Example 6 Identification of potential solubie surface and single pass membrane proteins 011 bovine sperm a) Purification ofbovine sperm 200 pl of extended bovine semen was loaded onto 2 ml 50% lTM PLUS column, and centrifugated at 1200 x g for 20 min at room temperature. 5 X 106 purified sperm cells resuspended in 1 ml of NCM (Table 2) were incubated with 10 ug/ml of biotinylated WGA (Vector Laboratories) for an hour at 28'C on a rotating platform. 25 pl ofwashed streptavidin DynabeadsTM (Invitrogen) were then incubated with the sperm for an hour at 28°C. DynabeadsTM/sperm complex was placed on magnet and washed three times with NCM. A minimal number of sperm was found in the supernatant, indicating that most sperm (>95%) formed a complex with the Dynabeads. b) e preparation for iTRAQ ng 500 pl of lysis buffer ning 7 M urea, 2 M ea, 4% CHAPS and 13 mM DTT was added to the DynabeadsTM/S X 106 sperm complex, vortexed, and incubated for 1 hr at 4C on a rotating platform. DynabeadsTM/sperm complex was then removed by magnet, and the supernatant was centrifugated at 14000 X g for 5 min to remove any remaining insoluble material. Protein estimation was performed using an EZQTM protein quantitation kit ular Probes). Filter—aided sample preparation method (FASP II) (Wisniewski et a1., 2009) was then used to prepare peptides from the sperm lysate.
Peptide s were added to 5% acetonitrile, and acidified with formic acid to pH 2 to 3. Samples were then desalted on Sep-Pak Vac tC18 solid phase extraction cartridge s), and completely dried in a vacuum concentrator. c! iTRAQ proteome analysis Dried peptide samples were sent to the APAF in Sydney for high sensitivity amino acid is and mass spectrometry analysis. Isobaric tags for relative and absolute quantitation (iTRAQ) was used for simultaneous identification and quantification of multiple peptide samples. 4—plex or 8-plex iTRAQ ts (SCIEX) were used to analyse 4 or 8 different biological s in a single experiment, respectively. In each iTRAQ ment, an equal amount of each peptide sample was labeled with a different iTRAQ reagent. Mixed iTRAQ-labeled peptide sample was then loaded onto a strong cation ion exchange (SCX) column and fractionated into 20 fractions. Each fraction was separated by reverse-phase gradient and injected into a Q-STAR Elite mass spectrometer. 2D-nanoLC-ESI~MS/MS data acquired was then analysed by ProteinPilot software 3.0 (AB1), and relative quantitation and protein identification were obtained. Paragon algorithm was used to perform database matching for n identification, protein grouping to remove abundant hits, and comparative quantitation. A thorough identification search was conducted in ProteinPilot. The IPI Bos taurus database (V3.49) was used for all searches. The data were normalized for loading error by bias correction using ProteinPilot. Proteins identified in multiple iTRAQ experiments were ed to each other, and also to the proteins identified in seminal plasma s. Ensembl genome browser (wvvwensembLorg) was used to check the presence of embrane domains and signal peptide sequence in each identified protein.
A list of 84 proteins in bovine sperm that may enhance sperm functionality and are likely to be on the sperm surface is provided in the Table 4. The sequences for these 84 proteins are provided in SEQ ID NO: 80-163, respectively. These proteins were selected from a total of 2206 ns identified across 19 different sperm lysates by the following criteria: unique proteins with a signal sequence that occurred in at least two experiments and that were not listed in Table 3, and additionally, proteins with known mitochondrial subcellular on or more than one transmembrane domain were omitted.
Table 4: Likely sperm surface proteins excluding seminal plasma proteins Protein descri ntion IPI identifier __Ensembl 1 rotein E) RDHll similar to retinol dehydrogenase 1 1 94814 ENSBTAP00000002535 isoform 1 GGH Gamma-imam 1 h drolase ENSBTAPOOOOOOO9917 PLBDl Putative whosholiase B-like 1 ENSBTAP00000020677 SCGBIDZ L AB 1P100842256 ENSBTAP00000044006 PLBDI PLBDl grotein IPIOO907129 ENSBTAP00000050579 TSBP TSBP rotein IP100840484 ENSBTAP00000001192 MGC165862 MGC16586213rotein IPIOO702545 P00000005987 NUP210L similar to nuclegpprin 2 lOkDa-like IP100705819 ENSBTAP00000006566 ZPBP Zona ellucida binding Eroiein 113100714900 ENSBTAP00000007229 LOC782909 similar to chromosome 9 open 19100702921 ENSBTAPOOOOOOO7827 readin' frame 79 1mm similar to izumo srrm—eg; fusion 1 113100701171 ENSBTAP00000015434 SPACAI Sperm acrosome membrane- IPIOO704953 ENSBTAP00000025934 associated rotein 1 CYBSRI NADH-c ochrome b5 reductase 1 IP100689803 ENSBTAP00000026548 D similar to Transmembrane protein IP100705075 ENSBTAP00000028044 LOC782834 o3 kD_a_Protein IP100709648 ENSBTAP00000031240 TSBP 63 kDa rotein IPIOO906483 ENSBTAP00000032083 BSG RPE7 rotein IPIOO696325 ENSBTAP00000039862 ENSBTAP00000041534 _-LOC10014I230 similar to chromosome 9 19100343355 ENSBTAP00000041933 P00000044123 LOC615968 r to Acrosome formation- IP100815450 ENSBTAP00000045709 associated factor _ ENSBTAP00000047419 PAM Peptidyl-glycine alpha-amidating 42571 ENSBTAP00000016466 monooxxgenase HTATIPZ HIV-1 Tat interactive protein 2, IPIOO760398 ENSBTAP00000017856 30kDa ADAM32 ADAM metallopeptidase domain 1P100707155 P00000031442 LOC786878 Uncharacterized protein IPIOO717926 ENSBTAP0000003 1847 C9orf134 homolo 1 ‘L _L_ _J CD46 11p cal LOC616002 20452 ENSBTAPOOOOOO41290 ADAM3A 83 kDapgotein ENSBTAP00000044565 LOC786599 similar to ADAM ‘ 07064 ‘ IP100823949 ENSBTAP00000046987 metallomtidase domain 20 preproprotein LOC530756 similar to acyltransferase like 1B lPIOO904OZ9 ENSBTAP00000048539 LYZL6 Lysozyme-likgpgotein 6 IPIOO715267 P00000000032 CRISP2 Cysteine-rich secretory_p_rotein 2 IP100699728 ENSBTAPOOOOOOOZSOS PPA2 P o nhos hatase anic) 2 IPIOO714601 ENSBTAP00000003 165 NUP155 similar to nucleoBorin lSSkDa IPIOO710810 ‘AP00000003 193 MFGE8 MFGES Brotein IP100689638 ENSBTAPOOOOOOO4272 VSTMQA MGC142894 rotein 111100694628 ENSBTAP00000004727 C13H200RF71 Short palate, lung and nasal IPIOO760496 ENSBTAP00000008942 e- ithelium oma ated 3Jlotein RUSCI 96 kDa rotein IPIOO904174 ENSBTAP00000009272 CPVL similar to Carboxypeptidase, IPIOO706544 ENSBTAP00000009404 Vitellogenic-like RNASE6 Ribonuclease K6 [P100702961 ENSBTAPOOOOOOI 1585 ADAMZ Disintegn'n and metalloproteinase IP100696982 ENSBTAP00000012384 domain-containin urotein 2 LYZL1 L so me-like -rotein1 IPI00696700 ENSBTAPO000001 3640 HBXA Beta-hexosaminidase subunit glpha 1P100702413 ENSBTAP00000017261 CPAl 89 kDa rotein 1P100843617 ENSBTAP00000017727 KLKBL4 KLKBL4 n IP100702428 P00000018804 - Pyruvate dehydrogenase phosphatase 1P100867405 ENSBTAP00000021895 re lato subunit (Reagent) PTI Pancreatic psin inhibitor 11100708836 ENSBTAP00000023042 LOC784519 similar to LOC512512 protein, 13657 ENSBTAP00000024347 ! artial LYZL4 L so c-like rotein4 IP100713792 P00000024756 - 39 kDa n GLI pathogenesis—relatedfl 87877 ENSBTAP00000025642 like 2 PLA2G7 Phosholiase A2, rou- VII IP100760435 ENSBTAP00000025719 IP100715275 ENSBTAP00000027467 HADHA FGF-Z bindin rotein [P100702650 ENSBTAP00000032860 ~ 26 kDa rotein IP10090647 l ENSBTAP00000033392 MGC137014 Hibernation-associated plasma 1P100689304 ENSBTAP00000037834 rotein HP—20-like APOB annoliorotein B IP100710056 LOC780846 Putative uncharacterized protein IPIOO686528 ENSBTAP00000041742 LOC784495 495 rotein [1’100829561 ENSBTAP00000042068 LOC614476 Putative acterlzed protein IP100694952 ENSBTAP00000043576 - 15 kDa protein IP100839329 ENSBTAP00000044687 ACRVI Acrosomal vesicle grotein 1 fliP100712714 LOC786289 similar to signal-regulatory 9100904540 P00000044718 rotein delta SPPl 31 kDa rotein 1P100840962 NME4 Non-metastatic cells 4, protein 93558 ENSBTAP000000451 10 exressed in — NDUFSG NDUFSQgrotein 1P100883392 ENSBTAP00000047840 LOC615258 similar to mCG4550 isoform 2 1_11’100906659 ENSBTAP00000050416 LOC780846 28 kDaflggein 1PIOO904088 ENSBTAPOOOOOOS 1 552 ] BSPHI 21 kDa Brotein IP100908264 ENSBTAP00000052231 SPAMl Seerm adhesion molecule 1 12321 ENSBTAP00000006089 PRCP Lysosomal Pro-X carboxypeplidase . 11000698864 ENSBTAP00000045060 TTR hfletin P100689362 BNSBTAPDOOOOO 14585 AGA As a l lucosaminjdase 1P100693170 ENSBTAP00000022716 HINTZ Histidine triad nucleotide—binding IP100689717 ENSBTAP00000015208 Ppiotein 2 ELSPBPl similar to epididymal sperm IP100700508 ENSBTAP00000021448 bindin urotein E12 CD59 CD59 molecule, ment regulatory IPIOO71 1804 ENSBTAP00000002967 protein Example 7 In vitro Sperm Testing A series of experiments are med in vitro to determine the y of a cholesterol-PEG-fimctional group conjugate to improve various measures of sperm functionality. Treated and untreated sperm are compared for changes in the following teristics: motility; membrane integrity; mitochondrial membrane potential; membrane fluidity; tin integrity; lipid peroxidation; capacitation; acrosome on; binding of antibodies, n and s to the sperm surface (or modified sperm surface proteins); ability of sperm to migrate in the FRT; the resistance of sperm to phagocytosis; and the ability of sperm to fertilize in vitro (see Table 5 for details).
Table 5: In vitro sperm testing CHARACTERISTIC :>(DU)<3»p< VJ REFERENCES Motility & (Tejerina et al., logy motility is for 10003 of 2008) cells. Can also indicate caacitation (h 1- ermotili ) Viability/ Flow try (FC)/ Depending upon the experiment, See (Gillan et Membrane integrity Fluorescent microscopy different vital dyes are used al., 2005) for a (FM) using a range of depending upon their properties review and dyes including (all available from Invitrogen). references Propidium iodide, Yo These dyes are used alone but within pro-l, Hoechst 33258 also in combination with other FC (H33258), assays described below. Overall LIVE/DEAD fixable far allows quantification of cells with red and SYBR 14 ermeant membranes Shape and FC/FM Enables quantification of size (Gillan et al., :4 anulari _1[ and cellular aggegation s 2005) _j Mitochondrial FC/FM with DilC1(5) The DilC,(5) dye is a member of (Garner et al., function/membrane (Invitrogen), J01 the cationic cyanine dyes that 1997; Shapiro potential (Invitrogen), rhodamine have been shown to accumulate in et al., 1979) 123 (Sigma) cells in response to membrane potential and thus permits quantification ofmitochondrial onality change. .101 and rhodamine 123 operates in a similar manner to DilC1(5) Capacitation status PC in combination with Capacitation induces sperm (Gadella and WGA-fluorescein surface changes. WGA/ Annexin Harrison, 2002; (Invitrogen)/ n V and merocyanin 540 all enable Mahmoud and V~fluorescein quantification of capacitation Parrish, 1996; /merocyanin 540 changes Medeiros and binding. Also the Parrish, 1996; y to undergo Rathi et al., acrosome reaction is 2001) used as a measure of caacitation Acrosorne integrity FC in combination with Both PNA and SBTI allow (Harper et al., 7 (lnvitrogen)/ quantification of changes on the 2008; Nagy et SBTI (So abean - 1- sin s-erm acrosome surface that al., 2003) TERISTIC ASSAY NOTES REFERENCES inhibitor)-488 reflect acrosome reaction, (Invitrogen) although this assay is generally used to r neous acrosome reaction. The ability of cells to acrosome react when initiated by calcium ionophore A23187 is also used as a measure of ca-acitation.
Surface antibody FC/FM in combination We have developed onal binding with antibodies antibodies to four seminal plasma proteins on bovine sperm (PDC- 109, BSP—A3, ESP—30 kDa, aSFP). These allow quantification of changes on the surface of nerrn Lectin binding FC/FM in ation Enables quantification of changes with lectins to sperm and sperm surface oroteins Heparin binding FC/FM with fluorescent Enables quantification of changes (Dapino et al., heparin (Invitrogen) in heparin binding to sperm and 2006) serm surface roteins Anandamide and Capacitation/motility/ Agonists and nists of (Gervasi et al., related compounds viability and acrosome inoid receptors (CB IR 2009; interaction with reaction and CBZR) modify sperm Maccarrone, sperm characteristics in vitro and may be 2009; involved in the regulation and Maccan'one et activation of ca . acitation al., 2005 ne fluidity PC with merocyanine Enables quantification of changes (Williamson et 540 in ne fluidi al., 1983) Assessment of Acridine orange sperm Enables quantification of (Ballachey et chromatin integrity chromatin structure chromatin integrity changes al., 1988; assay (SCSA; uses FC) L Evenson et al., 1980) Sperm ion Measure sperm Enables quantification of sperm (Aitken et al., assay in cervical migration in glass motility in a media that resembles 1992; Gillan et mucus capillary tube with at least part ofthe FRT al., 2008) fluorescent labeled sperm (H33342) Neutrophil Microscopic J‘Enables fication of sperm (Alghamdi et phagocytosis assay observation of phagocytosis by neutrophils al., 2009; phagocytes and Woelders and fluorescent labeled Matthijs, 2001) s with (H33342) Oviduct explants Binding of fluorescent Assays enable quantification of (Ignotz et al., binding/ Annexin-II labeled sperm (H33342) ability of sperm to bind to oviduct 2007; Teijeiro binding to oviduct. FC in or the likely receptor on the et at, 2009; combination with oviduct ki et al., fluorescent labeled 2005) Annexin—II protein Lipid dation FC/FM with C11- Enables fication of (Brouwers and BODIPY(58l/591) membrane lipid peroxidation Gadella, 2003) (lnvitrogen) Oxidative stress to FC with specific Enables quantification ofDNA (De luliis et al., - DNA antibody to 8-hydroxy- damage caused by oxidative 2009) xyguanosine stress in International ltd) ROS generation FC with Enablesgpantification ofROS (Bass et al., W0 2013/022354 2012/000140 CHARACTERISTIC REFERENCES Dihydroethidium (a species 1983; probe to detect Fridovich, superoxide) and 22,72— I997; Guthrie dichlorodihydro- and Welch, fluorescein 2006) diacetate (a probe to detect hydrogen a eroxide In vitro fertilization Do with titration of s overall quantification of (Amann and sperm the ability of sperm to bind zona Hammerstedt, pellucida, penetrate zona 2002; Lu and pellucida and fertilize the oocyte , 2004; Saeki et a1., 1995) Example 8 Sperm Maturation Model In this model, as detailed below, bovine sperm are incubated overnight in NCM under non-capacitating conditions (simulating the conditions sperm experience for the majority of the journey in the FRT, starting cell ity approximately 90%). Following overnight tion, sperm are diluted in capacitating media (CM; Table 6), triggering capacitation with high efficiency and minor loss of viability (cell viability in the 75-85% range). In typical experiments, when bovine sperm are capacitated with ne, db- cAMP and IBMX (3—isobutyl~1-methylxanthine), greater than 95% of viable cells capacitate as assessed by WGA-fluorescein/Annexin V or anine 540 binding (see Table 5; WGA staining is the most sensitive, with approximately 10-fold shift in the staining upon capacitation). When cells are capacitated in vitro they also gain the capacity to acrosome react (Table 5). Although the combination of caffeine, db-cAMP and IBMX is an nt inducer of capacitation, when more in Vivo like capacitation induction is required, heparin is used. Sperm treated with cholesterol-PEG—functional molecule are compared with untreated sperm for their ability to capacitate, in particular using heparin induction method.
Table 6: 1X CM itating media, pH 7.4) Component Concentration NaHzPO4 MgClz Sodium pyruvate 100mm so m: a) Day 1- Bovine sperm purification using PeroollTM PLUS A 90% PeroouTM PLUS solution is made by adding 10 X NCM to Person” PLUS. A 60% single layer gradient is then made by dilution with 1 X NCM. In the standard method, 4 ml of 60% PercollTM PLUS/NCM is added in a 15 m1 tube, 1.5 ml of ejaculate in liquid extender (standard tris-egg yolk, extension ~ 1:4 egg yolk-citrate- glycerol) is then gently loaded on top, and centrifuged at 700 X g for 20 min at room temperature. The pellet is d and washed once in 8 ml of NCM by centrifugation for 5 min at 700 X g. The supernatant is then removed and the pellet resuspended in 1 ml 1O of NCM. Capacitation ent tubes are set up at a sperm concentration of 5 X 107 cells/ml. b2 Day I - Flow cytometry analysis Samples are prepared for flow cytometry analysis as follows. The components shown in Table 7 below are incubated with 5 X 105 PercollTM PLUS-purified bovine sperm in a final volume of 200 pl at room temperature for 10 min, While propidium iodide (PI) is added just before ing by flow cytometry.
Table 7 FLUORESCENT FINAL 1 SUPPLIER COMPONENT CONCENTRATION Propidium iodide 03 uM Invitrogen x- PNA-alexa fluor 025 pg/ml T ogen i SBTI— alexa fluor 0.01 rig/ml l. Invitrogen WGA- fluorescein 0.00625 pig/ml ogen or WGA- alexa fluor 647 6) Day 1 - Incubation of bovine sperm sample overnight PercollTM PLUS-purified bovine sperm at 5 X 107 ml concentration are incubated in NCM overnight in a 28°C water bath. The sperm are then visually assessed under inverted bright field microscope and/or using QualiSperm prior to inducing capacitation. 6!) Day .2 — Transition of cells fiom non-capacitating media 1‘0 cagacitating media After overnight incubation, the cells are diluted in to CM (Table 2). Specifically, 1 ml of overnight incubated sperm is diluted 1:1 with 1 ml of CM media. Activators for capacitation, specifically caffeine and rib-CAMP are added at a final concentration of 1 mM (~ 16 hours afier tion started), and IBMX is added at a final concentration of 100 mM. Alternatively, bovine sperm capacitation is induced using heparin or methylbeta cyclodextrin (cholesterol acceptor). Samples are then incubated for an hour at 37°C. e) Day 2 - Flow cfiometly analysis of capacitated sperm r to day 1, bovine sperm samples are then incubated with fluorescently labeled SBTI, PNA and WGA for 10 min and PI added just prior to flow analysis.
In Vivo Field Artificial Insemination Trials Achieving pregnancy is dependent upon both the male and female fertility, and also upon other factors (such as management of animals, , age, environment, insemination procedure etc.) and thus analysis of male fertility y requires large numbers of animals in trials (Amann and Hammerstedt, 2002). At least for the bovine, the large number of sperm/ej aculate and also careful study design mean that many sources of variation can be controlled. In , Al trials have been conducted to look at number of sperm ed for insemination either alone (Den Daas et al., 1998) or in conjunction with other variables such as flow cytometry sorting (Bodmer et al., 2005), extender composition or other ation (Amann et al., 1999). The basic design is a sperm dose response using several bulls and a large number of cows (Den Daas et al., 1998).
In alternative studies, heterospermic inseminations with mixtures of treated and non-treated (control) sperm are employed to quickly determine fimctionality and/or fertility of the d sperm. In this experimental design, two distinguishable types of sperm are inseminated simultaneously, with the aim being to compare the different types of sperm and thus remove female fertility as an experimental variable. us reports have described heterospermic nation using sperm from multiple bulls (Dziuk, 1996; Flint et al., 2003), and a few s have been developed er with various techniques to assess the success ofthe sperm (Flint et al., 2003; Parrish and Foote, 1985).
In specific studies, semen is collected from a single bull and sperm are either treated with a cholesterol-PEG—fimctional molecule or left untreated. Treated and untreated ol) sperm are labelled with two different fluorescent dyes (such as Hoechst 33342 and Vybrant DyeCycle stains) to enable the Control and treated sperm to be distinguished. Equal s ofthe treated and control sperm are then siniultaneously inseminated into the same cow, and reciprocal studies are also carried out to ensure effects on sperm ort are not due to the marker fluorescent dye. Twelve to sixteen hours after spermic insemination the cow is slaughtered, the uterus and oviduct removed, and the ratio of treated and control sperm in the upper uterine horn and oviduct is determined. Significantly increased number of treated sperm are present in the upper uterine horn and oviduct compared to ted (control) sperm when the treatment successfiilly improves sperm functionality.
While the present invention has been described with reference to the c embodiments thereof, it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the invention. In addition, many modifications may be made to adapt a particular situation, material, composition of matter, method, method step or steps, for use in practicing the present invention. All such ations are intended to be within the scope of the claims appended hereto.
SEQ ID NO: 1463 are set out in the attached Sequence Listing. The codes for nucleotide sequences used in the attached Sequence Listing, including the symbol "n," conform to WIPO Standard ST.25 , Appendix 2, Table 1.
Bibliography Abuchowski, A., McCoy, J.R., Palczuk, N.C., van Es, T., and Davis, F.F. (1977a). Effect of covalent attachment of polyethylene glycol on genicity and circulating life of bovine liver catalase. J Biol Chem 252, 3582—3586.
Abuchowski, A., van Es, T., Palczuk, NC, and Davis, F.F. (1977b). Alteration of immunological properties of bovine serum albumin by covalent attachment of polyethylene glycol. J Biol Chem 252, 581.
, R.J., Bowie, H., Buckingham, D., Harkiss, 0., Richardson, D.W., and West, KM. (1992). Sperm penetration into a hyaluronic acid polymer as a means of ring functional competence. J Androl 13, 44-54.
Alghamdi, AS, and , D.N. (2005). Seminal DNase frees spermatozoa entangled in neutrophil extracellular traps. Biol Reprod 73, 1174-1181.
Alghamdi, A.S., Lovaas, B.J., Bird, S.L., Lamb, G.C., Rendahi, A.K., Taube, PC, and Foster, D.N. (2009). Species—specific interaction of seminal plasma on sperm-neutrophil binding. Anim Reprod Sci 114, 331-344.
Altschul, S.F., Gish, W., Miller, W., Myers, E.W., and Lipman, D.J. (1990). Basic local alignment search tool. J Mol Biol 215, 403-410.
Altschul, S.F., Madden, T.L., Schaffer, A.A., Zhang, J., Zhang, 2., Miller, W., and Lipman, D.J. (1997). Gapped BLAST and PSI—BLAST: a new generation of protein database search programs. Nucleic Acids Res 25, 3389-3402.
Alvarez, J.G., and Storey, ET. . Spontaneous lipid peroxidation in rabbit and mouse ymal spermatozoa: ence of rate on temperature and oxygen tration. Biol Reprod 32, 342-351.
Amann, R.P., and Hammerstedt, RH. (2002). Detection of differences in fertility. J Androl 23, 5.
Amann, R.P., Seidel, G.E., Jr., and Brink, Z.A. (1999). Exposure of thawed frozen bull sperm to a synthetic peptide before artificial insemination increases ity. J Androl 20, 4246. 3O Awda, B.J., Mackenzie—Bell, M., and Buhr, MM. (2009). Reactive Oxygen Species and Boar Sperm Function. Biol Reprod.
Ballachey, B.E., Evenson, D.P., and Saacke, R.G. . The sperm chromatin structure assay. Relationship with ate tests of semen quality and spermic performance of bulls. J Androl 9, 5.
Bass, D.A., Farce, J.W., Dechatelet, L.R., Szejda, P., Seeds, MC, and Thomas, M. (1983). Flow cytometric s of oxidative product formation by neutrophils: a graded response to ne stimulation. J lmmunol 130, 1910-1917.
Baumber, J., Ball, B.A., Gravance, C.G., Medina, V., and Davies-Morel, MC. (2000). The effect of reactive oxygen species on equine sperm motility, viability, acrosomal integrity, 4O mitochondrial membrane potential, and membrane lipid peroxidation. J Androl 21, 895- 902.
Bell, M, Sikka, S., Rajasekaran, M, and Hellstrom, W. (1992). Time course of hydrogen peroxide induced changes in the lipid peroxidation of human sperm membranes. Adv Contracept Deliv Syst 8, 144-150. 45 Belleannee, C., Belghazi, M., Labas, V., Teixeira-Gomes, A.P., Gatti, J.L., Dacheux, J.L., and Dacheux, F. (2011). Purification and identification of sperm surface proteins and changes during epididymal maturation. Proteomics 11, 1952-1964.
Bellin, M.E., Oyarzo, J.N., Hawkins, H.E., Zhang, H., Smith, R.G., Forrest, D.W., , LR, and Ax, R.L. (1998). Fertility-associated antigen on bull sperm indicates fertility 50 potential. J Anim Sci 76, 2032-2039.
Bienert, G.P., Schjoerring, J.K., and Jahn, T.P. (2006). Membrane transport of hydrogen peroxide. Biochim Biophys Acta 1758, 994—1003.
WO 22354 Bilodeau, J.F., Blanchette, 8., Cormier, N., and Sirard, MA. (2002). Reactive oxygen species—mediated loss of bovine sperm motility in egg yolk Tris extender: protection by pyruvate, metal chelators and bovine liver or oviductal fluid catalase. genology 57, 11054122.
, M., Janett, F., Hassig, M, den Daas, N., Reichert, P., and Thun, R. .
Fertility in heifers and cows after low dose insemination with sex-sorted and non-sorted sperm under field conditions. Theriogenology 64, 1647-1655.
Brouwers, J.F., and Gadella, BM. (2003). in situ detection and localization of lipid dation in individual bovine sperm cells. Free Radic Biol Med 35, 1382-1391.
Calvete, J.J., and Sanz, L. . insights into structure-function correlations of ungulate seminal plasma proteins. Soc Reprod Fertii Suppl 65, 5.
Chen, AM, and Scott, MD. (2001). Current and future ations of immunological attenuation via pegylation of cells and tissue. BioDrugs 15, 833-847.
Chen, 0., and Duan, E.K. . Aquaporins in sperm osmoadaptation: an emerging role for volume regulation. Acta Pharmacol Sin 32, 721-724.
Chen, (2., Peng, H., Lei, L., Zhang, Y., Kuang, H., Cao, Y., Shi, Q.X., Ma, T., and Duan, E. (2011). Aquaporin3 is a sperm water channel essential for postcopulatory sperm osmoadaptation and migration. Cell Res 21, 922-933.
D‘Amours, 0., Frenette, G., Fortier, M, Leclerc, P., and Sullivan, R. (2010). Proteomic comparison of detergent-extracted sperm proteins from bulls with different fertility indexes. Reproduction 139, 545-556.
Dapino, D.G., Marini, P.E., and Cabada, MO. (2006). Effect of heparin on in vitro tation of boar sperm. Biol Res 39, 9.
De luliis, G.N., Thomson, L.K., Mitchell, L.A., Finnie, J.M., Koppers, A.J., Hedges, A., Nixon, B., and Aitken, R.J. (2009). DNA damage in human spermatozoa is highly correlated with the efficiency of chromatin remodeling and the formation of 8-hydroxy-2'- uanosine, a marker of oxidative stress. Biol Reprod 81, 517-524. de Lamirande, E., and Gagnon, C. (1995). impact of reactive oxygen species on spermatozoa: a balancing act between beneficial and detrimental effects. Hum Reprod 10 Suppl 1, 15-21.
Den Daas, J.H., De Jong, G., Lansbergen, L.M., and Van Wagtendonk-De Leeuw, AM. (1998). The onship between the number of spermatozoa inseminated and the reproductive efficiency of individual dairy bulls. J Dairy Sci 81, 1714-1723.
Donev, RM, Sivasankar, 8., Mizuno, M, and Morgan, SP. (2008). The mouse complement regulator 0059b is significantly expressed only in testis and plays roles in sperm acrosome tion and motility. Moi lmmunol 45, 534642.
Drobnis, E.Z., and Overstreet, J.W. (1992). Natural history of mammalian spermatozoa in the female uctive tract. Oxt Rev Reprod Biol 14, 1-45.
Dziuk, P.J. (1996). Factors that influence the proportion of offspring sired by a male 4O following heterospermic insemination. Animal uction Science 43, 65-88.
Evenson, D.P., kiewicz, 2., and Melamed, MR. (1980). Relation of mammalian sperm tin heterogeneity to fertility. Science 210, 1131-1133.
Flesch, F.M., Brouwers, J.F., Nievelstein, P.F., Verkleij, A.J., van Goide, L.M., Colenbrander, B., and Gadella, BM. (2001). onate stimulated olipid 45 scrambling induces cholesterol redistribution and s cholesterol depletion in the sperm plasma membrane. J Cell Sci 114, 3543-3555.
Flint, A.F., Chapman, P.L., and Seidel, G.E., Jr. (2003). Fertility assessment through heterospermic insemination of flow-sorted sperm in cattle. J Anim Sci 81, 1814-1822.
Foote, RH. (2002). The history of artificial insemination: Selected notes and notables. 50 l of Animal Science 80, 1-10 Fraser, D.A., Harris, C.L., Williams, AS, Mizuno, M, Gallagher, 8., Smith, R.A., and Morgan, B.P. (2003). Generation of a recombinant, membrane-targeted form of the complement regulator 0059: activity in vitro and in vivo. J Biol Chem 278, 48921-48927.
Frenette, G., Girouard, J., and Sullivan, R. (2006). Comparison between epididymosomes collected in the intraluminal compartment of the bovine caput and cauda epididymidis. Biol Reprod 75, 0.
Fridovich, i. (1997). Superoxide anion radical (02-), xide dismutases, and related matters. J Biol Chem 272, 18515-18517. rs, A.C., Mullaart, E., Roelofs, R.M., van Hoorne, RP, Moreno, J.F., Moreno, 0., and Merton, J.S. (2009). What s fertility of sexed bull semen more, low sperm dosage or the sorting process? genology 71, 64-67.
Gadella, B.M., and Harrison, RA. . Capacitation induces cyclic ine 3',5'- monophosphate-dependent, but apoptosis-unrelated, exposure of aminophospholipids at the apical head plasma membrane of boar sperm cells. Biol Reprod 67, 340-350.
, D.L., Thomas, C.A., Joerg, H.W., tte, J.M., and Marshall, CE. (1997).
Fluorometric assessments of mitochondrial function and viability in eserved bovine spermatozoa. Biol Reprod 57, 1401—1406.
Gervasi, M.G., Rapanelii, M, Ribeiro, ML, , M., Bilii, 8., Franchi, AM, and Perez Martinez, 8. (2009). The endocannabinoid system in bull sperm and bovine oviductal epithelium: role of anandamide in sperm~oviduct interaction. Reproduction 137, 4.
Gillan, L., Evans, 6., and Maxwell, WM. (2005). Flow cytometric evaluation of sperm parameters in relation to fertility ial. Theriogenology 63, 445-457.
Gillan, L., Kroetsch, T., Maxwell, W.M., and Evans, G. (2008). Assessment of in vitro sperm characteristics in relation to fertility in dairy bulls. Anim Reprod Sci 103, 201-214.
Girouard, J., Frenette, G., and Sullivan, R. (2009). Compartmentalization of proteins in epididymosomes nates the association of epididymal proteins with the different functional structures of bovine spermatozoa. Biol Reprod 80, 965-972. ald, R.B., Choe, Y.H., e, J., and Conover, CD. (2003). Effective drug delivery by PEGylated drug conjugates. Adv Drug Deliv Rev 55, 217-250.
Guthrie, H.D., and Welch, GR. (2006). Determination of intracellular reactive oxygen species and high mitochondrial membrane ial in Percoll-treated viable boar sperm using fluorescence-activated flow cytometry. J Anim Sci 84, 2089-2100.
Harper, C.V., Cummerson, J.A., White, M.R., Publicover, S.J., and Johnson, PM. (2008). Dynamic resolution of acrosomal exocytosis in human sperm. J Cell Sci 121, 2130—2135.
Hawk, H.W. (1983). Sperm survival and transport in the female reproductive tract. J Dairy Sci 66, 26452660.
Hill, A., , S.H., Esser, D., Oldroyd, R.G., Cullen, M.J., Kareclas, P., Gallagher, 8., Smith, G.P., Richards, S.J., White, J., et al. (2006). Protection of erythrocytes from human complement-mediated lysis by membrane—targeted recombinant soluble CD59: new approach to PNH y. Blood 107, 2131-2137.
Hunter, RH. (2003). Advances in deep uterine insemination: a fruitful way fonivard to 4O t new sperm technologies in cattle. Anim Reprod Sci 79, 1574 70. lgnotz, G.G., Cho, M.Y., and Suarez, SS. (2007). Annexins are candidate oviductal receptors for bovine sperm surface proteins and thus may serve to hold bovine sperm in the oviductal reservoir. Biol Reprod 77, 906913.
Jackson, C.J., Charlton, J.L., Kuzminski, K., Lang, GM, and Sehon, AH. (1987). 45 Synthesis, isolation, and characterization of conjugates of ovalbumin with thoxypolyethylene glycol using cyanuric chloride as the coupling agent. Anal Biochem 165, 114-127.
Jevsevar, 8., Kunstelj. M., and Porekar, V.G. (2010). PEGylation of therapeutic proteins. hnol J 5. 113-128. 50 Johnson, L.A., Flook, JP, and Hawk, H.W. (1989). Sex preselection in rabbits: live births from X and Y sperm separated by DNA and cell sorting. Biol Reprod 41, 199—203.
Jones, R., Mann, T., and Sherins, R. (1979). Peroxidative breakdown of phospholipids in human spermatozoa, spermicidal properties of fatty acid peroxides, and protective action of seminal plasma. Fertil Steril 31, 531-537.
WO 22354 Katz, D.F., Drobnis, E.Z., and Overstreet, J.W. (1989). Factors regulating mammalian sperm migration through the female reproductive tract and oocyte vestments. Gamete Res 22, 443-469.
Kelly, V.C., Kuy, 8., Palmer, B.J., Xu, 2., Davis, SR, and Cooper, G.J. (2006).
Characterization of bovine seminal plasma by proteomics. Proteomics 6, 5826-5833.
Kirchhoff, C., and Hale, G. (1996). o-cell transfer of glycosylphosphatidylinositol- anchored membrane proteins during sperm maturation. Mol Hum Reprod 2, 177-184.
Kirchhoff, C., Pera, l., Derr, P., Yeung, C.H., and Cooper, T. (1997). The molecular y of the sperm surface. Post-testicular ne remodelling. Adv Exp Med Biol 1O 424, 221-232.
Klebanoff, S.J., and Smith, D.C. (1970). The source of H202 for the uterine fluid- mediated sperm-inhibitory system. Biol Reprod 3, 2.
Koppers, A.J., De luliis, G.N., Finnie, J.M., McLaughlin, EA, and Aitken, R.J. (2008).
Significance of mitochondrial reactive oxygen species in the generation of oxidative stress in spermatozoa. J Clin Endocrinol Metab 93, 3199-3207. te, 8., and Sirard, MA. (1998). Catalase and oviductal fluid reverse the decreased motility of bovine sperm in culture medium containing specific amino acids. J Androl 19, 31-36.
Lathrop, W.F., Carmichael, E.P., Myles, D.G., and Primakoff, P. (1990). cDNA cloning reveals the molecular structure of a sperm surface protein, PH-20, ed in sperm-egg adhesion and the wide distribution of its gene among mammals. J Cell Biol 111, 2939- 2949.
Lightfoot, R.J., and l, B.J. (1971). Effects of site of insemination, sperm motility and genital tract contractions on transport of spermatozoa in the ewe. J Reprod Fertil 26, 1- 13.
Lu, K.H., and Seidel, G.E., Jr. (2004). Effects of heparin and sperm concentration on cleavage and blastocyst development rates of bovine oocytes inseminated with flow cytometrically-sorted sperm. Theriogenology 62, 819—830.
Lutz, J.F., and Zarafshani, Z. (2008). Efficient construction of therapeutics, bioconjugates, biomaterials and bioactive es using azide—alkyne "click" chemistry.
Adv Drug Deliv Rev 60, 958-970.
Maccarrone, M. (2009). Endocannabinoids: friends and toes of reproduction. Prog Lipid Res 48, 344-354.
Maccarrone, M., Barboni, B., Paradisi, A., Bernabo, N., i, V., Pistilli, M.G., Fezza, F., Lucidi, P., and Mattioli, M. (2005). Characterization of the nnabinoid system ' boar spermatozoa and implications for sperm capacitation and acrosome reaction. J Cell Sci 118, 4393-4404.
Mahmoud, AL, and Parrish, J.J. (1996). Oviduct fluid and heparin induce similar surface changes in bovine sperm during capacitation: a flow cytometric study using lectins. Mol 40 Reprod Dev 43, 554—560. js, A., Engel, 8., and Woelders, H. (2003). Neutrophil recruitment and phagocytosis of boar spermatozoa after cial insemination of sows, and the effects of inseminate , sperm dose and ic additives in the extender. Reproduction 125, 357-367. 4-5 McCauley, TC, Zhang, H., Bellin, ME, and Ax, R.L. (1999). Purification and characterization of fertility-associated antigen (FAA) in bovine seminal fluid. Moi Reprod Dev 54, 3.
Medeiros, C.M., and Parrish, J.J. (1996). Changes in lectin binding to bovine sperm during n-induced capacitation. Mol Reprod Dev 44, 525-532. 50 Mitchell, J.R., Sanger, PL, and Rosenberger, J.L. (1985). Distribution and retention of spermatozoa with acrosomal and nuclear abnormalities in the cow genital tract. J Anim Sci 61, 956-967.
Moura, A.A., n, D.A., Koo, H., and Killian, G.J. (2006a). Proteins of the cauda ymal fluid associated with fertility of mature dairy bulls. J Androl 27, 534-541.
Moura, A.A., Chapman, D.A., Koo, H., and Killian, G.J. (2007). A comprehensive mic analysis of the accessory sex gland fluid from mature Holstein bulls. Anim Reprod Sci 98, 169-188.
Moura, A.A., Koc, H., Chapman, D.A., and Killian, G.J. (2006b). identification of proteins in the accessory sex gland fluid ated with fertility indexes of dairy bulls: a proteomic ch. J Androl 27, 201-211. s, K.J., and Saacke, R.G. (1989). Study of the functional anatomy of bovine al mucosa with special reference to mucus secretion and sperm transport. Anat Rec 225, 106-117.
NagY, 8., Jansen, J., Topper, E.K., and Gadeila, BM. (2003). A triple-stain flow cytometric method to assess plasma- and acrosome-membrane integrity of cryopreserved bovine sperm immediately after thawing in presence of egg-yolk particles.
Biol Reprod 68, 1828-1835.
Novak, 8., Ruiz-Sanchez, A., Dixon, W.T., Foxcroft, G.R., and Dyck, MK. (2010a).
Seminal plasma proteins as potential markers of relative fertility in boars. J Androl 31, 188-200.
Novak, S., Smith, T.A., Paradis, F., Burwash, L., Dyck, M.K., Foxcroft, GR, and Dixon, W.T. (2010b). Biomarkers of in vivo fertility in sperm and seminal plasma of fertile stallions. Theriogenology 74, 956-967.
Oren-Benaroya, R., Kipnis, J., and Eisenbach, M. . Phagocytosis of human post- capacitated spermatozoa by macrophages. Hum Reprod 22, 2947-2955.
Osheroff, J.E., Visconti, P.E., Valenzuela, J.P., Travis, A.J., z, J., and Kopf, GS. (1999). Regulation of human sperm capacitation by a cholesterol efflux-stimulated signal transduction pathway leading to protein kinase A-mediated up-regulation of protein tyrosine phosphorylation. Mol Hum Reprod 5, 1017-1026.
Otero, MO, and Nader-Macias, ME. (2006). inhibition of Staphylococcus aureus by H202—producing acillus gasseri isolated from the vaginal tract of cattle. Anim Reprod Sci 96, 35-46.
Parrish, J.J., and Foote, RH. . Fertility differences among male rabbits determined by spermic insemination of fluorochrome-labeled spermatozoa. Biol Reprod 33, 940949.
Rathi, R., Colenbrander, B., Bevers, MM, and Gadella, BM. (2001). Evaluation of in vitro capacitation of stallion spermatozoa. Biol Reprod 65, 462-470.
Roberts, M.J., Bentley, M.D., and Harris, JM. (2002). try for peptide and protein PEGylation. Adv Drug Deliv Rev 54, 459-476.
Rodriguez-Martinez, H., Kvist, U., Ernerudh, J., Sanz, L., and Calvete, J.J. (2011). l plasma proteins: what role do they play? Am J Reprod lmmunol 66 Suppl 1, 11- 22. , Saeki, K., Nagao, Y., Hoshi, M, and Nagai, M. (1995). Effects of heparin, sperm 4O concentration and bull variation on in vitro fertilization of bovine oocytes in a protein—free medium. Theriogenology 43, 751-759.
Salicioni, A.M., Piatt, M.D., Wertheimer, E.V., Arcelay, E., Allaire, A., Sosnik, J., and ti, PE. (2007). Signalling pathways involved in sperm capacitation. Soc Reprod Fertil Suppl 65, 245-259. 45 Schroter, S., Osterhoff, C., McArdle, W., and lvell, R. (1999). The glycocalyx of the sperm surface. Hum Reprod Update 5, 302-313.
Scott, M.D., Murad, K.L., Koumpouras, F., , M., and Eaton, J.W. . al lage of antigenic determinants: stealth erythrocytes. Proc Natl Acad Sci U S A 94, 7566-7571. 50 Senior, J., Delgado, 0., , 0., Tilcock, 0., and Gregoriadis, G. (1991). influence of surface hiiicity of liposomes on their ction with plasma protein and clearance from the circulation: studies with poly(ethylene glycol)-coated vesicles. Biochim Biophys Acta 1062, 77-82.
PCT/NZZOIZ/OOOMO Shannon, P., and Curson, B. (1972). Toxic effect and action of dead sperm on d bovine semen. J Dairy Sci 55, 0.
Shapiro, H.M., Natale, P.J., and Kamentsky, LA. (1979). Estimation of membrane potentials of individual lymphocytes by flow cytometry. Proc Natl Acad Sci U S A 76, 730. '3’ Sharpe, J.G., and Evans, KM. (2009). Advances in flow cytometry for sperm sexing. genology 71, 4-10.
Storey, B.T. (1997). Biochemistry of the induction and prevention of lipoperoxidative damage in human tozoa. Mol Hum Reprod 3, 203-213. 1O , ST. (2008). Mammalian sperm metabolism: oxygen and sugar, friend and toe. int J Dev Biol 52, 427437.
Sumner, J.B., and Dounce, A.L. (1937). Crystalline se. Science 85, 366—367.
Tang, 8., Garrett, C., and Baker, H.W. . Comparison of human cervical mucus and artificial sperm penetration media. Hum Reprod 14, 2812-2817.
Teijeiro, J.M., lgnotz, G.G., and Marini, PE. (2009). Annexin A2 is involved in pig (Sus scrofa)sperm—oviduct interaction. Moi Reprod Dev 76, 334-341.
Tejerina, F., Buranaamnuay, K., Saravia, F., Wallgren, M., and uez-Martinez, H. (2008). Assessment of motility of ated, liquid-stored boar spermatozoa using computerized instruments. Theriogenology 69, 1129-1 138.
Teramura, Y., and lwata, H. (2009). Surface modification of islets with PEG-lipid for improvement of graft survival in intraportai transplantation. Transplantation 88, 624-630.
Tollner, T.L., Venners, S.A., Hollox, E.J., Yudin, A.l., Liu, X., Tang, 6., Xing, H., Kays, R.J., Lau, T., Overstreet, J.W., et al. (2011). A Common Mutation in the Defensin DEFB126 Causes Impaired Sperm Function and Subtertility. Sci Transl Med 3, 92ra65.
Tollner, T.L., Yudin, A.l., Tarantal, A.F., Treece, C.A., Overstreet, J.W., and Cherr, G.N. (2008a). Beta-defensin 126 on the surface of e sperm mediates attachment of sperm to oviductal epithelia. Biol Reprod 78, 400-412.
Tollner, T.L., Yudin, A.t., Treece, C.A., Overstreet, J.W., and Cherr, G.N. (2008b).
Macaque sperm coating protein DEF8126 facilitates sperm penetration of cervical mucus. Hum Reprod 23, 2523—2534.
Topfer-Petersen, E., Romero, A., Varela, P.F., Ekhlasi-Hundrieser, M, ova, Z., Sanz, L., and Calvete, J.J. . Spermadhesins: a new n family. Facts, hypotheses and perspectives. Andrologia 30, 217-224. .
Tosic, J., and Walton, A. (1950). Metabolism of spermatozoa. The formation and elimination of en peroxide by spermatozoa and effects on motility and survival.
Biochem J 47, 2. z, J.M., Martinez, E.A., Roca, J., Gil, MA, Parrilla, l., Cuello, C., Carvaial, G., Lucas, X., and Vazquez, J.L. (2005). improving the efficiency of sperm technologies in pigs: the value of deep intrauterine insemination. Theriogenology 63, 7. 4O Vazquez, J.M., Ross, J., Gil, MA, Cuello, C., Parrilla, l., Vazquez, J.L., and Martinez, EA. (2008). New developments in low-dose insemination technology. Theriogenology 70, 1216-1224.
Vernet, P., Fulton, N., Wallace, 0., and Aitken, R.J. (2001). Analysis of reactive oxygen species generating systems in rat epididymal spermatozoa. Biol Reprod 65, 1102-1113. 45 ti, P.E., Ning, X., Fornes, M.W., Alvarez, J.G., Stein, P., Connors, S.A., and Kopf, GS. (1999). Cholesterol efflux-mediated signal transduction in mammalian sperm: cholesterol release signals an increase in protein ne phosphorylation during mouse sperm capacitation. Dev Biol 214, 429-443.
Waberski, D., Magnus, F., Mendonca Ferreira, F., Petrunkina, A.M., Weitze, K.F., and 50 Tooter-Petersen, E. (2005). Importance of sperm-binding assays for fertility prognosis of porcine spermatozoa. Theriogenology 63, 4.
Williamson, P., ks, K., and el, RA. (1983). Merocyanine 540, a fluorescent probe sensitive to lipid packing. Biochim Biophys Acta 732, 387-393.
Wisniewski, J.R., Zougman, A., Nagaraj, N., and Mann, M. (2009). Universal sample ation method for proteome analysis. Nat Methods 6, 359-362.
Woelders, H., and js, A. (2001). ytosis of boar spermatozoa in vitro and in vivo. Reprod Suppl 58, 113-127.
Yeung, CH. (2010). Aquaporins in spermatozoa and testicular germ cells: identification and potential role. Asian J Androi 12, 490-499.
Yudin, A.l., Tollner, T.L., Li, MW, Treece, CA, reet, J.W., and Cherr, G.N. (2003). ESP13.2, a member of the efensin family, is a macaque sperm surface— coating protein involved in the capacitation process. Biol Reprod 69, 1118-1128. 1D Yudin, A.l., Treece, C.A., Tollner, T.L., Overstreet, J.W., and Cherr, SN. (2005). The carbohydrate structure of DEFB126, the major component of the lgus Macaque sperm plasma membrane glycocalyx. J Membr Biol 207, 119—129.
Zalipsky, 8., Brandeis, E., Newman, M.S., and Woodle, MC. (1994). Long circulating, cationic liposomes containing amino—PEG—phosphatidylethanolamine. FEBS Lett 353, 71-74.
We

Claims (7)

claim:
1. A conjugate sing: (a) a membrane anchoring agent comprising a lipid; (b) polyethylene glycol; and (c) a functional molecule that is effective in improving the onality of a cell by attachment of said functional molecule to the cell surface of said cell, wherein the functional molecule is selected from the group consisting of: proteins, carbohydrates and biotin, and is attached to the polyethylene glycol by an attachment group.
2. The conjugate of claim 1, wherein said cell is a sperm and wherein said improvement is increasing the lifespan of said sperm in the female reproductive tract.
3. The conjugate of claim 1 or 2, wherein the onal molecule is selected from the group consisting of: se; glutathione; CD55; CD59; CD73; DNaseI; SPAMl; and functional ts thereof.
4. The conjugate of claim 1 or 2, n the functional molecule is a polypeptide comprising an amino acid sequence selected from the group consisting of: SEQ ID NO: 1-163; and functional variants thereof.
5. The conjugate of any one of claims 1 to 4, wherein the attachment group is selected from the group consisting of: ester amine reactive ; maleimide; vinyl sulfone; iodoacetamide; orthopyridyl disulfide; hydrazide; benzotriazole; succinimidyl carbonate; and succinimidyl active esters based on onic and ic acids.
6. The conjugate of any one of claims 1 to 5, wherein the lipid is selected from the group consisting of: cholesterol, diacylglycerolipids, dialkylglycerolipids, glycerophospholipids, sphingosine derived diacyl- and dialkyl- lipids, ceramide, phosphatidate, phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl , phosphatidyl inositol and phosphatidyl glycerol.
7. The conjugate of any one of claims 1 to 6, wherein the conjugate comprises a structure of any one of
NZ620290A 2011-08-11 2012-08-08 Methods and compositions for improving sperm functionality NZ620290B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161522609P 2011-08-11 2011-08-11
US61/522,609 2011-08-11
PCT/NZ2012/000140 WO2013022354A1 (en) 2011-08-11 2012-08-08 Methods and compositions for improving sperm functionality

Publications (2)

Publication Number Publication Date
NZ620290A NZ620290A (en) 2015-03-27
NZ620290B2 true NZ620290B2 (en) 2015-06-30

Family

ID=

Similar Documents

Publication Publication Date Title
US20140315185A1 (en) Methods and compositions for improving sperm functionality
US8592138B2 (en) Methods for improving sperm functionality
Plante et al. Evolution and function of mammalian binder of sperm proteins
Lapointe et al. Binding of a bovine oviductal fluid catalase to mammalian spermatozoa
Spencer et al. Insights into conceptus elongation and establishment of pregnancy in ruminants
Muiño‐Blanco et al. Seminal plasma proteins and sperm resistance to stress
Georgiou et al. Modulation of the oviductal environment by gametes
AU2012351977B2 (en) Stimulation of ovarian follicle development and oocyte maturation
Srivastava et al. Bovine seminal PDC-109 protein: An overview of biochemical and functional properties
Rego et al. Seminal plasma protein profiles of ejaculates obtained by internal artificial vagina and electroejaculation in Brahman bulls
Arangasamy et al. Isolation and characterization of heparin and gelatin binding buffalo seminal plasma proteins and their effect on cauda epididymal spermatozoa
Marzoni et al. A proteomic approach to identify seminal plasma proteins in roosters (Gallus gallus domesticus)
Argento-Cerü et al. Localization of diamine oxidase in animal tissues
Ijaz et al. In vitro‐cultured bovine granulosa and oviductal cells secrete sperm motility‐maintaining factor (s)
WO2011108946A1 (en) Methods for improving sperm functionality
Mogielnicka-Brzozowska et al. Proteome of cat semen obtained after urethral catheterization
BRPI0706690A2 (en) composition and process for enhancing mammalian seminal function
Maňásková et al. Localization of porcine seminal plasma (PSP) proteins in the boar reproductive tract and spermatozoa
NZ620290B2 (en) Methods and compositions for improving sperm functionality
Dobrinski et al. Isolation and characterization of a protein with homology to angiotensin converting enzyme from the periacrosomal plasma membrane of equine spermatozoa
Iwamoto et al. Influence of seminal plasma components on sperm motility
Bhatt et al. Fertilization, embryonic development and oviductal environment: role of estrogen induced oviductal glycoprotein
Kutty et al. An overview on binder of sperm proteins of cattle.
Dutta et al. Differential influence of recombinant non-glycosylated and glycosylated glycodelin on human sperm function: comparative studies with hamster spermatozoa
WO2024048795A1 (en) Ruminant conception chance-improving composition