US20090011496A9 - Enhancement of mammalian embryo development - Google Patents

Enhancement of mammalian embryo development Download PDF

Info

Publication number
US20090011496A9
US20090011496A9 US11/370,152 US37015206A US2009011496A9 US 20090011496 A9 US20090011496 A9 US 20090011496A9 US 37015206 A US37015206 A US 37015206A US 2009011496 A9 US2009011496 A9 US 2009011496A9
Authority
US
United States
Prior art keywords
embryos
embryo
prostaglandin
iloprost
blastocyst
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US11/370,152
Other versions
US7709259B2 (en
US20060160213A1 (en
Inventor
Jaou-Chen Huang
Jennifer Goldsby
Wan-Song Wun
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Texas System
Original Assignee
Jaou-Chen Huang
Goldsby Jennifer S
Wun Wan-Song A
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to EP04783425A priority Critical patent/EP1670902A4/en
Priority to PCT/US2004/029167 priority patent/WO2005026324A2/en
Application filed by Jaou-Chen Huang, Goldsby Jennifer S, Wun Wan-Song A filed Critical Jaou-Chen Huang
Priority to US11/370,152 priority patent/US7709259B2/en
Publication of US20060160213A1 publication Critical patent/US20060160213A1/en
Publication of US20090011496A9 publication Critical patent/US20090011496A9/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF TEXAS HLTH SCI CTR HOUSTON
Assigned to BOARD OF REGENTS OF THE UNIVERSITY OF TEXAS SYSTEM reassignment BOARD OF REGENTS OF THE UNIVERSITY OF TEXAS SYSTEM ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HUANG, JAOU-CHEN, WUN, WAN-SONG A., GOLDSBY, JENNIFER S.
Application granted granted Critical
Publication of US7709259B2 publication Critical patent/US7709259B2/en
Expired - Fee Related legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0604Whole embryos; Culture medium therefor
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/02Compounds of the arachidonic acid pathway, e.g. prostaglandins, leukotrienes

Definitions

  • the present invention generally relates to compositions and methods for in vitro culturing of mammalian embryos and for enhancing achievement of pregnancy after implantation of the cultured embryo in the uterus of a suitable mammalian host. More particularly, the invention pertains to such composition and methods which employ a prostaglandin or prostaglandin analog for enhancing blastocyst quality and improving implantation potential of a treated embryo.
  • a method of enhancing in vitro development of an embryo comprising supplementing culture medium with a prostaglandin such as PGI 2 or PGE 2 or analog thereof.
  • the prostaglandin is PGI 2 or PGE 2
  • the prostaglandin analog is Iloprost or PGE 1 .
  • the method includes adding the prostaglandin or analog to the culture medium in an amount sufficient to promote complete hatching of the embryo. Preferably “complete hatching” comprises freeing the embryo from the zona pellucida.
  • supplementing of the culture medium is done during early developmental stage of the embryo during which time the genome is activated and the fertilized egg develops into morulae.
  • the embryo in which the embryo is human, the early developmental stage commences with a four-to eight-cell state.
  • supplementing of the culture medium is done during the morulae to early blastocyst stage development of the embryo.
  • supplementing of the culture medium is performed during both the early developmental stage to morulae stage development of the embryo and during the morulae to early blastocyst stage development of the embryo.
  • the supplementing step includes exposing the embryo in culture to the prostaglandin or analog between 24 to 42 hours after harvest of the embryo at its two-cell developmental stage.
  • the step of supplementing during the morulae to early blastocyst stage comprises exposing the embryo to prostaglandin or prostaglandin analog at a time between 42 to 72 hours after harvest of the embryo at its two-cell developmental stage.
  • “Implantation potential” is the ability of the embryos to implant in the uterus. This method includes carrying out one of the above-described embodiments to enhance in vitro development of the embryo, such that complete hatching of the embryo in culture is achieved or hatching is enhanced, compared to other IVF methods.
  • the completely or nearly-hatched embryo is then introduced into the uterus of a mammalian host, such than enhanced implantation of the embryo is achieved.
  • complete hatching of the embryo in vitro correlates with establishment of a viable pregnancy.
  • Live birth potential refers to the ability of an embryo to yield a live birth.
  • the method comprises enhancing in vitro development of the embryo, as described above, by in vitro culturing with an amount of a prostaglandin or analog thereof, such that enhanced hatching potential or complete hatching of the embryos in culture is achieved.
  • such treatment confers an enhanced potential for complete hatching even after the embryos are removed from culture and are in an in vivo environment.
  • the hatched embryo, or the embryo with enhanced hatching potential is then transferred to the uterus of a mammalian host; and the embryo is allowed to implant and grow in vivo, such that the ability of the embryo to yield a live birth is enhanced relative to that of an embryo that is implanted without enhancement of hatching.
  • a method of increasing the live birth rate in a group of in vitro fertilized mammalian embryos comprises enhancing in vitro development of the embryos according to an above-described method, such that enhancement of hatching, or, preferably, complete hatching of the embryos in culture is achieved.
  • the method includes introducing or transferring the embryos into mammalian hosts; and then allowing the embryos to grow in vivo, wherein the rate of live births from the group of embryos with enhanced hatching is greater than the rate of live births from the group of embryos transferred into a host uterus without first being completely hatched or receiving enhancement of hatching.
  • an improved cell culture medium for in vitro development of a mammalian embryo wherein the improvement includes a supplemental amount of a prostaglandin or analog thereof effective to enhance hatching, and, preferably, to promote complete hatching of the embryo in vitro.
  • the medium contains an amount of prostaglandin, or analog thereof, that is effective to enhance complete hatching potential of the embryo after it has been removed from in vitro culture and transferred to the uterus of a mammalian host.
  • an in vitro method of enhancing the development of a mammalian embryo to at least the blastocyst stage includes obtaining an in vitro fertilization embryo comprising at least two cells; and growing the embryo in a culture medium supplemented with an amount of a prostaglandin or a prostaglandin analog effective for enhancing the quality of a resulting blastocyst.
  • enhancing blastocyst quality comprises blastocyst size.
  • enhancing blastocyst quality comprises increasing inner cell mass of the blastocyst.
  • enhancing blastocyst quality comprises increasing at least two qualities chosen from the group consisting of blastocyst size, inner cell mass and trophectoderm size.
  • the prostaglandin is prostacyclin (PGI 2 ) or prostaglandin E 2 (PGE 2 ).
  • the prostaglandin analog comprises a prostacyclin agonist (e.g., Iloprost).
  • the prostaglandin analog comprises prostaglandin E 1 (PGE 1 ).
  • Also provided in accordance with the present invention is a method of increasing the in vivo implantation potential of an in vitro fertilization embryo, which comprises culturing a mammalian zygote in vitro in a culture medium supplemented with an amount of a prostaglandin or a prostaglandin analog effective to promote an increase in quality of resulting blastocysts, whereby the in vivo implantation potential of a resulting embryo is enhanced.
  • culturing of a mammalian zygote in a culture medium supplemented with the prostaglandin or prostaglandin analog further enhances establishment of a viable pregnancy when the resulting embryo is implanted in a uterus.
  • FIG. 1 is a graph showing prostacyclin (PGI 2 ) and complete hatching of mouse embryos.
  • PGI 2 prostacyclin
  • Two-cell mouse embryos were cultured with PGI 2 analog (Iloprost, 1 nM to 10 ⁇ M). The rate of complete hatching was determined 96 hours later. Results from three to five independent experiments (17-20 embryos each) are expressed as mean ⁇ S.D. The ED 50 value is approximately 6.7 nM.
  • FIG. 2 is a bar graph showing augmentation of complete hatching and duration of prostacyclin (PGI 2 ) exposure (Iloprost exposure based on stages of embryo development).
  • PGI 2 prostacyclin
  • Two-cell mouse embryos were cultured in the presence of 0.1 ⁇ M Iloprost, a PGI 2 analog, during the indicated periods (expressed as hours after the harvest of two-cell embryos).
  • the rate of complete hatching is expressed as mean ⁇ S.D.
  • the developmental stages during which the embryos were exposed to Iloprost are listed in the table. Two periods appear critical to the full effects of Iloprost: 24-42 hours and 42-72 hours, corresponding to the development from eight-cell embryos to morulae and morulae to early blastocysts, respectively.
  • FIG. 3 is a Western blot showing expression of prostacyclin receptor (IP) in mouse embryos.
  • IP prostacyclin receptor
  • FIGS. 4 A-E and G are photomicrographs showing expression of prostacyclin receptor (IP) in morulae and blastocysts.
  • Phase contrast FIG. 4A
  • IP staining FIG. 4B
  • FIG. 4C shows propidium iodide staining of nuclei in a blastocyst.
  • FIG. 4D shows IP and propidium iodide staining of the blastocyst are superimposed.
  • FIG. 4E shows IP staining of the blastocyst shows a reticular pattern.
  • FIG. 4F is a sketch showing the location of the inner cell mass (ICM) within the blastocyst.
  • FIG. 4G is the confocal microscopy image of a blastocyst showing that only the trophectoderm is stained by IP antibody. The plane of the section is indicated by the line in the panel F. There was no IP staining in negative controls, unfertilized oocytes or embryos at other developmental stages (not shown). The bar in the figure is approximately 20 ⁇ m.
  • FIG. 5 is a bar graph showing Iloprost binding to intact mouse embryos.
  • One hundred sixteen mouse blastocysts were incubated with 0.1 ⁇ M 3 H-Iloprost, a PGI 2 analog, in the presence or absence of 5 ⁇ M unlabeled Iloprost. Bound and free 3 H-Iloprost were separated by membrane filtration.
  • FIGS. 6 A-B show gestation sacs in the uterus of gestational carriers. Implantation was determined by an increase in vascularity at the implantation site and confirmed by the presence of gestation sacs. Seventy two hours after embryo transfer, the gestational carrier was given 1% Chicago Blue via the tail vein and euthanized three minutes later.
  • FIG. 6A shows discrete blue bands (indicated by *) encircling the uterine horn that received embryos.
  • each blue band corresponded to a gestation sac, which appeared as pink-colored tissue in the opened uterine horn. No blue bands or gestation sacs were found in the uterine horn that did not receive embryos (marked by an arrow).
  • the present disclosure developed in the course of investigating the effects of PGI 2 and PGE 2 on sperm and the embryo, and in subsequent investigations.
  • Our recent results indicate that human [Huang, 2002] and mouse [Huang, 2004a] oviductal epithelial cells express enzymes essential to the synthesis of PGI 2 , i.e. COX-1 or COX-2 and PGI 2 synthase.
  • Abundant PGI 2 and PGE 2 was produced when 14 C-arachidonic acid was incubated with microsomes prepared from human [Huang, 2002] or mouse oviduct [Huang, 2004a].
  • the present study also considered the fact that sperm travel to distal oviduct to fertilize the egg and the first 72 hours of embryo development takes place in the oviduct.
  • mice were kept under controlled temperature, humidity, and light cycle (12 hour light/dark cycle) conditions with free access to water and food.
  • Three-week old C57B1/6 female mice were purchased from Harlan (Indianapolis, Ind., USA). Eight-week old C3H male mice were purchased initially from Harlan and later from The Jackson Laboratory (Bar Harbor, Me., USA).
  • Superovulation in the female mice was achieved by intraperitoneal injection of pregnant mare serum gonadotropin (5 IU), followed by human chorionic gonadotropin (hCG, 5 IU) 46 hours later. After receiving hCG, each female mouse was paired with one fertile male mouse. Forty-eight hours later, two-cell embryos were harvested from the oviduct into ⁇ -MEM media supplemented with 25 mM HEPES and 1% BSA (Irvine Scientific).
  • Embryos (17-20 per group) were cultured at 37° C. under 5% CO 2 in a four-well dish (Nalge Nunc International, Naperville, Ill., USA) containing 600 ⁇ l of medium in each well.
  • the HTF and the ⁇ -MEM media were used sequentially during the 96-hour period to meet the changing nutritional requirements of the cleaving embryos [Gardner, 1998].
  • the HTF medium SAGE Biopharma
  • the ⁇ -MEM medium Irvine Scientific
  • the experimental embryos received Iloprost in water and the control embryos received an equal amount of water.
  • Immunoreactive protein was detected by incubation with the antibody and visualization with enhanced chemi-fluorescence (Amersham Biosciences, Piscataway, N.J., USA), detected using a STORM 860 laser scanner (Amersham Biosciences). Human platelet microsomes were used as positive controls. The antibody specificity was confirmed in parallel experiments using pre-absorbed antibody.
  • Mouse embryos were fixed in 4% paraformaldehyde (pH 7.4) at 4° C. for 30 min. After three washes in PBS, the embryos were blocked for 20 min at room temperature in Tris-buffered saline (pH 7.4) containing 0.05% Tween-20, 5% powdered milk and 0.1% Triton X-100. The embryos were incubated with IP antibody (5 ng/ml) in blocking buffer for 2 hours then with goat anti-rabbit IgG coupled with Alexa 488 (2.5 ⁇ g/ml, Molecular Probes, Eugene, Oreg., USA) for 30 min at 37° C.
  • IP antibody 5 ng/ml
  • Alexa 488 2.5 ⁇ g/ml
  • Fluorescence microscopy was performed using a Zeiss AxioPlan 2 microscope (Carl Zeiss, Baden-Wuerttemberg, Germany) equipped with appropriate filters. Images were captured using a CCD camera and processed by the AxioVision program (Version 3.0.6). Confocal microscopy was performed using a BioRad Radiance 2000 confocal system (Bio-Rad Laboratories, Hercules, Calif., USA) attached to an Olympus BX-50 microscope. The images were processed using the Image-Pro + program (Media-Cybernetics, Carlsbad, Calif., USA).
  • the reaction was terminated by transferring the blastocysts to 2 ml of ice-cold wash buffer (0.01% BSA in 10 mM HEPES, pH 7.4).
  • the buffer and the blastocysts were filtered through glass fiber filters (Whatman GF/C 2.4 cm) and the filters were washed three times with 2 ml of wash buffer.
  • the filters were dried in an oven before the radioactivity was determined by scintillation counting with 5 ml of scintillation fluid.
  • vaginal smears were obtained from potential gestational carriers (eight week old C3B6F1 female mice) to determine the stage of estrus cycle. Those in estrus were paired with vasectomized ICR males (Harlan) and checked for vaginal plugs on the following morning. Those with plugs were designated to be 0.5 day pseudopregnant.
  • Embryo transfer was performed on 2.5 day of pseudopregnancy under a dissecting microscope (Olympus SZ-PT). Surgical anesthesia was obtained through intraperitoneal injection of ketamine (200 mg/Kg) and xylazine (10 mg/Kg) (both from Burns Vet Supply Inc. TX, USA). Each uterine horn was accessed via a 2-cm flank incision. With the proximal oviduct held by a pair of forceps, an opening was created at the distal end of the uterine horn on the anti-mesenteric side with a 30 gauge needle. The opening permits the entry of transfer pipette which has an inner diameter of 135 ⁇ m (MidAtlantic Diagnostics, Inc. NJ, USA).
  • implantation was determined based on a previously described method with some modifications (Paria et al., 1993). Briefly, three minutes before euthanasia, 0.1 ml Chicago Blue (1%) was injected via the tail vein of the gestational carrier. After the carrier was sacrificed, the uterine horns were removed. Discrete blue bands (indicated by *) encircling the uterine horns were counted ( FIG. 6A ). The presence of these bands reflect the increased vascular supply to the implantation sites. The uterine horns were then opened and the gestation sacs counted ( FIG. 6B ). The implantation rate was expressed as the number of gestation sacs per embryo transferred.
  • gestational carriers cannibalize their pups when the litter size was small.
  • gestational carriers 14 days after embryo transfer (16.5 days of pregnancy or two days before natural birth). After euthanasia, the number of live pups was counted and the weight of individual pup and placenta was determined. All pups were examined for gross anomalies. The number of empty gestation sacs was also counted in order to determine the implantation rate.
  • the live birth rate was expressed as the number of live pups per embryo transferred; the implantation rate was expressed as the number of sacs, with or without live pups, per embryo transferred.
  • the Fisher's exact test was used to compare the rates; the Student's t-test was used to compare the weights. A p ⁇ 0.05 was considered as statistically significant.
  • the GraphPad Instat® software (GraphPad Software Inc., San Diego, Calif., USA) was used for statistical analysis.
  • oviduct-derived PGI 2 is complementary to endometrium-derived PGI 2 , which mediates endometrial decidualization to ensure receptivity [Lim, 1999].
  • the enhanced hatching mediated by PGI 2 or PGE 2 may be due to an increased number of embryonic cells.
  • Different mechanisms are involved in the hatching of mouse embryo in vitro and in vivo. Hatching in vitro involves blastocyst expansion, causing a global zonal thinning prior to zonal rupture, whereas hatching in vivo involves global zonal lysis by uterine or trophectodermal lysins [Montag, 2000]. Therefore, a sufficiently high number of embryonic cells is required to accomplish hatching in vitro.
  • PGI 2 enhanced the complete hatching of cultured mouse embryos.
  • the stage-specific expression of IP by the mouse embryos coincided with their responsiveness to PGI 2 .
  • PGE 2 also enhanced the complete hatching of cultured mouse embryos.
  • Table 1 shows that PGI 2 enhanced the implantation potentials of mouse embryos. Eighty-four control embryos and 81 Iloprost-treated embryos were transferred to 12 gestational carriers. Seventy two hours later, more gestation sacs were found in the Iloprost-treated group than the control group (76% versus 42%, relative risk 1.84, 95% confidence interval 1.38-2.43, p ⁇ 0.0001). At the time of transfer, the developmental stages of the embryos were comparable.
  • Table 2 shows that PGI 2 Iloprost enhanced the live birth potentials of mouse embryos.
  • the live birth rates of the control and the experimental groups were 28% and 36%, respectively (p 0.0017, relative risk 1.28, 95% confidence interval: 1.044-1.560).
  • PGI 2 increased the potential for live births of mouse embryos without affecting the weight of either the pups or the placentae. In addition, no gross anomalies were noted.
  • PGI 2 initiates a chain of events culminating in enhanced hatching, implantation, and increased live births.
  • PAF platelet-activating factor
  • They may include increased embryonic cell number [Montag, 2000], augmented production of trypsin-like proteases by the trophectoderm [Sawada, 1990; Perona, 1986], enhanced blastocoele expansion due to enhanced Na + -K + -ATPase system of the trophectoderm [Biggers, 1988] or yet to be discovered mechanism(s).
  • control embryos were comparable (42% and 46%) when examined 72 hours or 14 days after transfer, respectively.
  • IVF culture media with substances that enhance embryo development or implantation has been adopted by most laboratories and media manufacturers with great care.
  • the concerns may include issues regarding reproductive toxicology, teratogenecity, and possibly, epigenetic effects of such manipulation.
  • Lambs from ovine embryos cultured in media supplemented with human serum were heavier and had a longer gestation period [Thompson, 1995; Holm, 1996; Sinclair, 1999].
  • PGI 2 enhances the potentials of implantation and live births of mouse embryos without affecting the weights of either the fetuses or the placentae, i.e., prostacyclin did not increase the weights of the placenta or the baby.
  • Previously reported agents which enhance implantation and live births also increase the weight of the babies, making those agents unsuitable for human in vitro fertilization (IVF) use.
  • prostacyclin did not affect the weights of the placenta and the babies in the in vivo mouse model, it can be considered for use in human IVF embryos.
  • mice embryos are considered representative or predictive of similar favorable in vivo results that will be obtained in other mammalian embryos, including human, when those embryos are similarly treated in vitro with prostaglandin or a prostaglandin analog prior to being transferred to the uterus.
  • the current human IVF involves the transfer of 8-cell stage or blastocyst stage embryos to the patient's uterus.
  • the human IVF is similar to the design of our experiment involving transferring control or prostacyclin analog-treated embryos to gestational carriers.
  • Our data showed, when transferred to gestational carriers, experimental embryos exposed to prostacyclin analog between 8-cell and morula stages had significantly higher rates of implantation and live births than control embryos (as shown in Tables 1 and 2, respectively).

Abstract

A method of enhancing in vitro development of a mammalian embryo is disclosed which comprises supplementing the culture medium with a prostaglandin, or a prostaglandin analog, in an amount effective to promote complete hatching of the embryo (i.e., freeing of the embryo from the zona pellucida). The quality of human blastocysts is enhanced in vitro by culturing with a prostacyclin agonist, Iloprost. The in vivo implantation potential and live birth potential of an in vitro fertilization embryo is thereby enhanced and establishment of a viable pregnancy is facilitated.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation-in-part of PCT/US04/029167 filed Sep. 8, 2004, from which priority under 35 U.S.C. §120 is claimed. This application also claims the benefit under 35 U.S.C. § 119(e) of U.S. Provisional Patent Application No. 60/708,291 filed Aug. 15, 2005. The disclosures of those applications are hereby incorporated herein by reference.
  • STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
  • This invention was made in part, with funding from the National Institute of Child Health and Human Development (NICHD), Contract No. HD01277. Accordingly, the United States Government has certain rights in this invention.
  • BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • The present invention generally relates to compositions and methods for in vitro culturing of mammalian embryos and for enhancing achievement of pregnancy after implantation of the cultured embryo in the uterus of a suitable mammalian host. More particularly, the invention pertains to such composition and methods which employ a prostaglandin or prostaglandin analog for enhancing blastocyst quality and improving implantation potential of a treated embryo.
  • 2. Description of the Related Art
  • The environment within the oviduct enhances the fertilization potential of sperm and promotes the development of cleaving embryos. Embryos co-cultured with oviductal epithelial cells have been shown to have improved development, hatching and implantation [Xu, 2001; Xu, 2000]. Prostacyclin (PGI2) is traditionally thought to be involved in maintaining blood and vascular homeostasis. However, recent observations in gene knock out mice suggest that it may have other physiological functions. The importance of endometrium-derived PGI2 is underscored by the observations made in the cyclooxygenase (COX)-2 knockout mice [Lim, 1997]. The endometrium of the COX-2 knock out mice failed to decidualize normally. As a result, transferred wild type embryos could not implant in the female mice deficient in COX-2. The aforementioned abnormalities could be, to some extent, corrected by administration of an exogenous PGI2 analog to the mother [Lim, 1999].
  • The role of oviduct-derived PGI2 in the development of pre-implantation embryos is not clear, because the litter size of PGI2 receptor (IP) knockout mice has not been compared with that of the wild type [Murata, 1997]. However, the genotypic distribution of pups arising from mating heterozygous IP knockout mice failed to conform to Mendel's Law: the prevalence of male and female pups with homozygous IP knockout genotype were 37% and 20%, respectively, less than expected [Murata, 1997].
  • SUMMARY OF PREFERRED EMBODIMENTS
  • It was recently discovered that the human oviduct synthesizes abundant prostacyclin (PGI2) and PGE2 [Huang, 2002]. Gene knockout studies suggest that endometrium-derived PGI2 is essential to endometrial decidualization, but the effects of PGI2 on the development of pre-implantation embryos have not been reported. Employing the hypothesis that optimal development of preimplantation embryos might require PGI2, the effects of PGI2 on mouse embryos were examined based on the rates of complete hatching. The expression of PGI2 receptor (IP) was evaluated by Western blot analysis and immunohistochemistry. The binding of PGI2 to embryos was confirmed by radioligand binding assay. Our results demonstrate that the hatching of mouse embryos is enhanced by supplementation of Iloprost (ED50 6.7 nM), a stable PGI2 synthetic analog. Exposure to Iloprost during eight-cell to morulae or morulae to early blastocyst stages was critical to enhanced hatching. This stage-specific response to PGI2 coincided with the developmental stage-specific expression of IP. Further studies were conducted in an attempt to correlate the enhanced embryo hatching with increased implantation and live births, mouse embryos cultured in media supplemented with Iloprost were transferred to gestational carriers. The number of gestation sacs and live pups was compared with that of control embryos. Because additives in culture media reportedly increased fetal weights [DeBaun, 2002; Thompson, 1995; Sinclair, 1999], the weights of the pups and the placentae were also compared. Our results indicate that culturing embryos in media supplemented with Iloprost enhances the implantation and live birth of mouse embryos without affecting the weights of the pups or the placentae.
  • Accordingly, in certain embodiments of the present invention, a method of enhancing in vitro development of an embryo comprising supplementing culture medium with a prostaglandin such as PGI2 or PGE2 or analog thereof is provided. In some embodiments, the prostaglandin is PGI2 or PGE2, and in some embodiments the prostaglandin analog is Iloprost or PGE1. In some embodiments, the method includes adding the prostaglandin or analog to the culture medium in an amount sufficient to promote complete hatching of the embryo. Preferably “complete hatching” comprises freeing the embryo from the zona pellucida. In some embodiments of the method, supplementing of the culture medium is done during early developmental stage of the embryo during which time the genome is activated and the fertilized egg develops into morulae. In some embodiments in which the embryo is human, the early developmental stage commences with a four-to eight-cell state. In some embodiments of the method, supplementing of the culture medium is done during the morulae to early blastocyst stage development of the embryo. In some embodiments, supplementing of the culture medium is performed during both the early developmental stage to morulae stage development of the embryo and during the morulae to early blastocyst stage development of the embryo. In certain embodiments of the method, in which the embryo is of mouse origin, the supplementing step includes exposing the embryo in culture to the prostaglandin or analog between 24 to 42 hours after harvest of the embryo at its two-cell developmental stage. In certain embodiments of the method wherein the embryo is mouse, the step of supplementing during the morulae to early blastocyst stage comprises exposing the embryo to prostaglandin or prostaglandin analog at a time between 42 to 72 hours after harvest of the embryo at its two-cell developmental stage.
  • Also provided in accordance with certain embodiments of the present invention, is a method of increasing the in vivo implantation potential of an in vitro fertilization embryo. “Implantation potential” is the ability of the embryos to implant in the uterus. This method includes carrying out one of the above-described embodiments to enhance in vitro development of the embryo, such that complete hatching of the embryo in culture is achieved or hatching is enhanced, compared to other IVF methods. In accordance with certain embodiments of this method, the completely or nearly-hatched embryo is then introduced into the uterus of a mammalian host, such than enhanced implantation of the embryo is achieved. In some embodiments, complete hatching of the embryo in vitro correlates with establishment of a viable pregnancy.
  • In some embodiments of the present invention, a method of increasing the live birth potential of an in vitro fertilized mammalian embryo is provided. “Live birth potential” refers to the ability of an embryo to yield a live birth. The method comprises enhancing in vitro development of the embryo, as described above, by in vitro culturing with an amount of a prostaglandin or analog thereof, such that enhanced hatching potential or complete hatching of the embryos in culture is achieved. In preferred embodiments such treatment confers an enhanced potential for complete hatching even after the embryos are removed from culture and are in an in vivo environment. This property is referred to as “enhanced hatching potential.” The hatched embryo, or the embryo with enhanced hatching potential, is then transferred to the uterus of a mammalian host; and the embryo is allowed to implant and grow in vivo, such that the ability of the embryo to yield a live birth is enhanced relative to that of an embryo that is implanted without enhancement of hatching.
  • In yet another embodiment of the present invention, a method of increasing the live birth rate in a group of in vitro fertilized mammalian embryos is provided which comprises enhancing in vitro development of the embryos according to an above-described method, such that enhancement of hatching, or, preferably, complete hatching of the embryos in culture is achieved. The method includes introducing or transferring the embryos into mammalian hosts; and then allowing the embryos to grow in vivo, wherein the rate of live births from the group of embryos with enhanced hatching is greater than the rate of live births from the group of embryos transferred into a host uterus without first being completely hatched or receiving enhancement of hatching.
  • Still further provided in accordance with certain embodiments of the invention is an improved cell culture medium for in vitro development of a mammalian embryo wherein the improvement includes a supplemental amount of a prostaglandin or analog thereof effective to enhance hatching, and, preferably, to promote complete hatching of the embryo in vitro. In some embodiments the medium contains an amount of prostaglandin, or analog thereof, that is effective to enhance complete hatching potential of the embryo after it has been removed from in vitro culture and transferred to the uterus of a mammalian host.
  • In accordance with certain embodiments, an in vitro method of enhancing the development of a mammalian embryo to at least the blastocyst stage is provided. The method includes obtaining an in vitro fertilization embryo comprising at least two cells; and growing the embryo in a culture medium supplemented with an amount of a prostaglandin or a prostaglandin analog effective for enhancing the quality of a resulting blastocyst. In some embodiments, enhancing blastocyst quality comprises blastocyst size. In some embodiments, enhancing blastocyst quality comprises increasing inner cell mass of the blastocyst. In some embodiments, enhancing blastocyst quality comprises increasing at least two qualities chosen from the group consisting of blastocyst size, inner cell mass and trophectoderm size. In some embodiments, the prostaglandin is prostacyclin (PGI2) or prostaglandin E2 (PGE2). In some embodiments, the prostaglandin analog comprises a prostacyclin agonist (e.g., Iloprost). In some embodiments, the prostaglandin analog comprises prostaglandin E1 (PGE1).
  • Also provided in accordance with the present invention is a method of increasing the in vivo implantation potential of an in vitro fertilization embryo, which comprises culturing a mammalian zygote in vitro in a culture medium supplemented with an amount of a prostaglandin or a prostaglandin analog effective to promote an increase in quality of resulting blastocysts, whereby the in vivo implantation potential of a resulting embryo is enhanced. In some embodiments, culturing of a mammalian zygote in a culture medium supplemented with the prostaglandin or prostaglandin analog further enhances establishment of a viable pregnancy when the resulting embryo is implanted in a uterus. These and other embodiments, features and advantages of the present invention will become apparent with reference to the following description and drawings.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a graph showing prostacyclin (PGI2) and complete hatching of mouse embryos. Two-cell mouse embryos were cultured with PGI2 analog (Iloprost, 1 nM to 10 μM). The rate of complete hatching was determined 96 hours later. Results from three to five independent experiments (17-20 embryos each) are expressed as mean±S.D. The ED50 value is approximately 6.7 nM.
  • FIG. 2 is a bar graph showing augmentation of complete hatching and duration of prostacyclin (PGI2) exposure (Iloprost exposure based on stages of embryo development). Two-cell mouse embryos were cultured in the presence of 0.1 μM Iloprost, a PGI2 analog, during the indicated periods (expressed as hours after the harvest of two-cell embryos). The rate of complete hatching is expressed as mean±S.D. The developmental stages during which the embryos were exposed to Iloprost are listed in the table. Two periods appear critical to the full effects of Iloprost: 24-42 hours and 42-72 hours, corresponding to the development from eight-cell embryos to morulae and morulae to early blastocysts, respectively. Exposure to Iloprost during either period ensured enhanced hatching. The numbers of independent observations (each with 18-20 embryos) were: control, 12; 42-72 hours, 3; others, 4. The rate of complete hatching in the control embryos was different from that in FIG. 1, probably because the source of the male mice was changed.
  • FIG. 3 is a Western blot showing expression of prostacyclin receptor (IP) in mouse embryos. Western blot analysis of total cell lysate from 60 mouse blastocysts (lane 1) and microsomes of human platelets (positive control, lane 2) showed immunoreaction with antibody against human IP. The migration of mouse IP was less than that of human IP, consistent with the expected molecular weights of ˜50 kDa and ˜46 kDa, respectively.
  • FIGS. 4A-E and G are photomicrographs showing expression of prostacyclin receptor (IP) in morulae and blastocysts. Phase contrast (FIG. 4A) and IP staining (FIG. 4B) images of two morulae are shown. The zona pellucida is indicated by the arrow. FIG. 4C shows propidium iodide staining of nuclei in a blastocyst. In FIG. 4D the IP and propidium iodide staining of the blastocyst are superimposed. FIG. 4E shows IP staining of the blastocyst shows a reticular pattern. Clustered IP staining is seen only in the trophectoderm; the inner cell mass shows no increased staining. FIG. 4F is a sketch showing the location of the inner cell mass (ICM) within the blastocyst. FIG. 4G is the confocal microscopy image of a blastocyst showing that only the trophectoderm is stained by IP antibody. The plane of the section is indicated by the line in the panel F. There was no IP staining in negative controls, unfertilized oocytes or embryos at other developmental stages (not shown). The bar in the figure is approximately 20 μm.
  • FIG. 5 is a bar graph showing Iloprost binding to intact mouse embryos. One hundred sixteen mouse blastocysts were incubated with 0.1 μM 3H-Iloprost, a PGI2 analog, in the presence or absence of 5 μM unlabeled Iloprost. Bound and free 3H-Iloprost were separated by membrane filtration.
  • FIGS. 6A-B show gestation sacs in the uterus of gestational carriers. Implantation was determined by an increase in vascularity at the implantation site and confirmed by the presence of gestation sacs. Seventy two hours after embryo transfer, the gestational carrier was given 1% Chicago Blue via the tail vein and euthanized three minutes later. FIG. 6A shows discrete blue bands (indicated by *) encircling the uterine horn that received embryos. In FIG. 6B, each blue band corresponded to a gestation sac, which appeared as pink-colored tissue in the opened uterine horn. No blue bands or gestation sacs were found in the uterine horn that did not receive embryos (marked by an arrow).
  • DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS
  • The present disclosure developed in the course of investigating the effects of PGI2 and PGE2 on sperm and the embryo, and in subsequent investigations. Our recent results indicate that human [Huang, 2002] and mouse [Huang, 2004a] oviductal epithelial cells express enzymes essential to the synthesis of PGI2, i.e. COX-1 or COX-2 and PGI2 synthase. Abundant PGI2 and PGE2 was produced when 14C-arachidonic acid was incubated with microsomes prepared from human [Huang, 2002] or mouse oviduct [Huang, 2004a]. The present study also considered the fact that sperm travel to distal oviduct to fertilize the egg and the first 72 hours of embryo development takes place in the oviduct.
  • As part of the present study, the impact of PGI2 on the embryos was evaluated by observing the complete hatching of mouse embryos cultured with PGI2 analog. To further elucidate the mechanism, the expression of IP and the binding of embryos by radio-labeled Iloprost were also studied.
  • Materials and Methods
  • Source of Reagents and Institutional Approval
  • Unless stated otherwise, reagents were purchased from Sigma Co. (St. Louis, Mo., USA). The care of the laboratory animals and the research protocols were approved by the Animal Welfare Committee. Use of human samples was approved by the Committee for the Protection of Human Subjects (equivalent to an Institutional Review Board). These human embryo studies were carried out without funding from the United States Government.
  • Harvest and Culture of Mouse Embryos.
  • Mice were kept under controlled temperature, humidity, and light cycle (12 hour light/dark cycle) conditions with free access to water and food. Three-week old C57B1/6 female mice were purchased from Harlan (Indianapolis, Ind., USA). Eight-week old C3H male mice were purchased initially from Harlan and later from The Jackson Laboratory (Bar Harbor, Me., USA). Superovulation in the female mice was achieved by intraperitoneal injection of pregnant mare serum gonadotropin (5 IU), followed by human chorionic gonadotropin (hCG, 5 IU) 46 hours later. After receiving hCG, each female mouse was paired with one fertile male mouse. Forty-eight hours later, two-cell embryos were harvested from the oviduct into α-MEM media supplemented with 25 mM HEPES and 1% BSA (Irvine Scientific).
  • Embryos (17-20 per group) were cultured at 37° C. under 5% CO2 in a four-well dish (Nalge Nunc International, Naperville, Ill., USA) containing 600 μl of medium in each well. The HTF and the α-MEM media were used sequentially during the 96-hour period to meet the changing nutritional requirements of the cleaving embryos [Gardner, 1998]. The HTF medium (SAGE Biopharma) was used during the first 48 hours, and the α-MEM medium (Irvine Scientific), with Earle's Salts and 2 mM glutamine, was used during the second 48 hours. The experimental embryos received Iloprost in water and the control embryos received an equal amount of water. Preliminary experiments showed that more than 95% of the embryos became blastocysts by 96 hours, similar to those cultured in KSOM media (Specialty Media, Cell and Molecular Technologies, Inc. Phillipsburg, N.J., USA). After 96-hour culture, each embryo was examined for the presence of the zona pellucida. Embryos completely free of the zona pellucida were counted as having completely hatched. The rate of complete hatching was determined by dividing the number of completely hatched embryos by the total number of embryos. Complete hatching of embryos was chosen as an endpoint instead of blastocyst formation or embryo hatching because the latter two markers are not correlated with establishment of a viable pregnancy [Lane, 1997].
  • Western Blot Analysis.
  • The deduced amino acid sequences of mouse IP (417 a.a., Genebank_BAA05144) and human IP (386 a.a., Genebank_BAA06110) are highly homologous [Katsuyama, 1994]. Preliminary studies confirmed that an affinity-purified polyclonal peptide antibody (a gift from Dr. Ke-He Ruan, University of Texas Health Science Center) cross-reacted with mouse IP. Western blot analysis was performed as described previously [Huang, 2002]. Briefly, total cell lysate from 60 mouse blastocysts was separated by electrophoresis on a 10% acrylamide gel (PAGE) and transferred to a nitrocellulose membrane (Schleicher & Schuell, Inc., Keene, N.H., USA). Immunoreactive protein was detected by incubation with the antibody and visualization with enhanced chemi-fluorescence (Amersham Biosciences, Piscataway, N.J., USA), detected using a STORM 860 laser scanner (Amersham Biosciences). Human platelet microsomes were used as positive controls. The antibody specificity was confirmed in parallel experiments using pre-absorbed antibody.
  • Immunohistochemistry, Fluorescence Microscopy and Confocal Microscopy.
  • Mouse embryos were fixed in 4% paraformaldehyde (pH 7.4) at 4° C. for 30 min. After three washes in PBS, the embryos were blocked for 20 min at room temperature in Tris-buffered saline (pH 7.4) containing 0.05% Tween-20, 5% powdered milk and 0.1% Triton X-100. The embryos were incubated with IP antibody (5 ng/ml) in blocking buffer for 2 hours then with goat anti-rabbit IgG coupled with Alexa 488 (2.5 μg/ml, Molecular Probes, Eugene, Oreg., USA) for 30 min at 37° C. Cell nuclei were counterstained with 10 μg/ml propidium iodide at room temperature for 20 min. The embryos were mounted in Fluoromount-G® (Southern Biotechnology Associates Inc., Birmingham, Ala., USA). For fluorescence microscopy, blastocysts were placed in the mounting media and overlaid with a coverslip. For confocal microscopy, a spacer of approximately 50 μm was placed between the slide and the coverslip to maintain the three dimensional morphology of the embryo. For negative controls, embryos were incubated with 10 ng/ml non-immune rabbit IgG (i.e., twice the concentration of primary antibody).
  • Fluorescence microscopy was performed using a Zeiss AxioPlan 2 microscope (Carl Zeiss, Baden-Wuerttemberg, Germany) equipped with appropriate filters. Images were captured using a CCD camera and processed by the AxioVision program (Version 3.0.6). Confocal microscopy was performed using a BioRad Radiance 2000 confocal system (Bio-Rad Laboratories, Hercules, Calif., USA) attached to an Olympus BX-50 microscope. The images were processed using the Image-Pro+ program (Media-Cybernetics, Carlsbad, Calif., USA).
  • Whole Embryo Radioligand Binding Assay.
  • Analysis of the binding of 3H-Iloprost to blastocysts was performed as previously described [Arbab, 2002] with slight modification. Two hundred thirty-two hatched and hatching blastocysts were washed three times in binding buffer (10 mM MnCl2 in 10 mM HEPES, pH 7.4) and then transferred to 100 μl of binding buffer with or without unlabeled Iloprost (5 μM). The reaction was started by adding an equal amount of binding buffer containing 3H-Iloprost (200 nM, specific activity 11.0 Ci/mmol, Amersham Biosciences) and incubating for 60 min at room temperature. The reaction was terminated by transferring the blastocysts to 2 ml of ice-cold wash buffer (0.01% BSA in 10 mM HEPES, pH 7.4). The buffer and the blastocysts were filtered through glass fiber filters (Whatman GF/C 2.4 cm) and the filters were washed three times with 2 ml of wash buffer. The filters were dried in an oven before the radioactivity was determined by scintillation counting with 5 ml of scintillation fluid.
  • Statistical Analysis
  • Student's t-test or one-way analysis of variance followed by Dunnett test were used where appropriate. A p<0.05 was considered statistically significant. Construction of dose response curves and calculation of ED50 values (with the Hill slope set at 1.0) were completed with the GraphPad Prism® software (GraphPad Prism Software Inc., San Diego, Calif., USA).
  • Embryo Transfer
  • Mouse embryos were harvested and cultured as described above, and subsequently published by the present inventor (Huang et al., 2003). Briefly, two-cell embryos (C3B6 μl) were harvested from super-ovulated, three-week old C57B1/6 female mice 42 hours after hCG injection. The embryos (14 per group) were cultured at 37° C. under 5% CO2 in four-well dish (Nalge Nunc International, Naperville, Ill., USA) each containing 600 μl HTF media (SAGE Biopharma, Bedminster, N.J., USA). The experimental embryos received Iloprost (1 μM, Caymen Chemical, Ann Arbor, Mich., USA) in water and the control embryos, an equal amount of water. In either case, the volume of water was less than 0.1% of the culture media.
  • The day after the embryo donors received hCG, vaginal smears were obtained from potential gestational carriers (eight week old C3B6F1 female mice) to determine the stage of estrus cycle. Those in estrus were paired with vasectomized ICR males (Harlan) and checked for vaginal plugs on the following morning. Those with plugs were designated to be 0.5 day pseudopregnant.
  • Embryo transfer was performed on 2.5 day of pseudopregnancy under a dissecting microscope (Olympus SZ-PT). Surgical anesthesia was obtained through intraperitoneal injection of ketamine (200 mg/Kg) and xylazine (10 mg/Kg) (both from Burns Vet Supply Inc. TX, USA). Each uterine horn was accessed via a 2-cm flank incision. With the proximal oviduct held by a pair of forceps, an opening was created at the distal end of the uterine horn on the anti-mesenteric side with a 30 gauge needle. The opening permits the entry of transfer pipette which has an inner diameter of 135 μm (MidAtlantic Diagnostics, Inc. NJ, USA). Up to seven embryos in 0.8 μl of transfer media (α MEM with 25 mM HEPES and 1% BSA) were transferred to each horn. After each transfer, contents of the pipette were examined under a stereo microscope to identify retained embryos. The flank incision was closed with 9 mm metallic clips (Clay Adams, Parsippany, N.J., USA). To avoid mixing of control and experimental embryos due to embryo migration from one horn to the other (personal communication with Dr. Andreas Zimmer, University of Bonn, Germany), each gestational carrier received either control or experimental embryos. To maintain consistent transfer techniques, the embryo transfer was performed according to the same protocol and by one individual, who was blinded to the treatment the embryos being placed received. To ensure that both groups benefited equally from the experience gained over the course of the study (˜6 months), the embryos were assigned treatments (control or Iloprost) in blocks of four.
  • Determination of Implantation Rate
  • Seventy-two hours after embryo transfer, implantation was determined based on a previously described method with some modifications (Paria et al., 1993). Briefly, three minutes before euthanasia, 0.1 ml Chicago Blue (1%) was injected via the tail vein of the gestational carrier. After the carrier was sacrificed, the uterine horns were removed. Discrete blue bands (indicated by *) encircling the uterine horns were counted (FIG. 6A). The presence of these bands reflect the increased vascular supply to the implantation sites. The uterine horns were then opened and the gestation sacs counted (FIG. 6B). The implantation rate was expressed as the number of gestation sacs per embryo transferred.
  • Determination of Live Birth Rate and the Weights of Pups and Placentae
  • Preliminary studies showed some gestational carriers cannibalize their pups when the litter size was small. In order to count and weigh the pups accurately, we sacrificed gestational carriers 14 days after embryo transfer (16.5 days of pregnancy or two days before natural birth). After euthanasia, the number of live pups was counted and the weight of individual pup and placenta was determined. All pups were examined for gross anomalies. The number of empty gestation sacs was also counted in order to determine the implantation rate. The live birth rate was expressed as the number of live pups per embryo transferred; the implantation rate was expressed as the number of sacs, with or without live pups, per embryo transferred.
  • Statistical Analysis
  • The Fisher's exact test was used to compare the rates; the Student's t-test was used to compare the weights. A p<0.05 was considered as statistically significant. The GraphPad Instat® software (GraphPad Software Inc., San Diego, Calif., USA) was used for statistical analysis.
  • Results
  • PGI2 Enhanced the Complete Hatching of Mouse Embryos
  • The complete hatching of mouse embryos was enhanced by Iloprost in a concentration-dependent manner. The effects were statistically significant at 0.1 μM or higher (FIG. 1). Maximum augmentation of complete embryo hatching occurred at 1 μM, where 81±7% (mean±S.D. n=3) of the experimental embryos hatched completely. In contrast, only 49±14% (mean±S.D. n=5) of the control embryos hatched completely. The ED50 value derived from the dose response curve was 6.7 nM (FIG. 1). The saturable, concentration-dependent responses and the ED50 value of 6.7 nM suggest that the effects of Iloprost were mediated by a receptor.
  • Duration of PGI2 Exposure and the Developmental Stages of Embryos Critical to the Enhanced Hatching.
  • Because embryos undergo several developmental stages before entering the uterus, we investigated developmental stage-specific responses to Iloprost. Our results indicate that some periods during the 96-hour culture were more critical than others. Exposure to Iloprost during the first 24 hours of culture (during this period most of the two-cell embryos developed into eight-cell embryos) did not enhance hatching (FIG. 2). On the other hand, exposure to Iloprost between 0-72 hours or 24-72 hours after harvest yielded the same rates of complete hatching as did 0-96 hours exposure (FIG. 2).
  • The two critical periods of exposure to Iloprost that ensured the full effects of Iloprost were between 24 to 42 hours and 42 to 72 hours after harvest, corresponding to the transformation from eight-cell embryos to morulae and from morulae to early blastocysts, respectively (table at bottom of FIG. 2). Our data indicate that brief exposure of embryos to PGI2 confers an enhanced potential for complete hatching even after the embryos leave the oviduct.
  • Mouse Embryos Express IP.
  • To investigate the developmental stage-specific expression of IP in mouse embryos, we performed Western blot analysis on blastocysts and immunohistochemistry on embryos at different developmental stages. Western blot analysis showed a protein of the expected molecular weight was detectable by the affinity-purified antibody against IP (FIG. 3). Fluorescence microscopy showed IP staining was present in morulae and blastocysts (FIG. 4A-E) but not in unfertilized egg, or in one-cell, two-cell, four-cell and eight-cell embryos (not shown). The IP staining in morulae and blastocysts shared the same fine, reticular pattern. Confocal microscopy images suggest that IP was preferentially expressed in the trophectoderm (FIG. 4G). Thus, the stage-specific expression of IP by the mouse embryo coincided with the responsiveness to Iloprost.
  • Radiolabeled Iloprost Binds to Mouse Embryo.
  • The concentration-dependent enhancement of complete embryo hatching by Iloprost suggested that the effect was receptor-mediated. We performed a radioligand binding assay to further confirm the binding of mouse embryos by Iloprost. We elected to quantitate Iloprost binding at a 0.1 μM level of the ligand, where augmented hatching was first observed and found that approximately 3.04 fmol 3H-Iloprost bound specifically to 116 blastocysts (FIG. 5).
  • Discussion
  • Our data indicate, for the first time, that PGI2 enhances complete hatching of embryos. This concurs with previous reports that co-culture of mouse embryos with epithelial cells from human oviducts increased embryo cell number, reduced apoptosis, and improved embryo hatching [Piekos, 1995; Xu, 2000; Xu, 2001]. Thus, oviduct-derived PGI2 and vascular endothelium-derived PGI2 exert their effects in a similar, paracrine fashion. The former enhances embryo hatching; the latter prevents platelet aggregation. The concentration-dependent response was consistent with a receptor-mediated event and the ED50 value of 6.7 nM was similar to the reported Kd value of solublized IP (8 nM) from human platelets [Tsai, 1989].
  • The response of embryos to Iloprost was developmental stage-specific and coincided with the expression of IP (FIG. 2). These critical periods for responsiveness coincided with the sojourn of mouse embryos in the oviduct, during which time the fertilized eggs develop into morulae. The relevant developmental stages also coincided with the activation of genome in human and mouse embryos, which takes place between the four- and eight-cell stages and after the two-cell stage, respectively [Tesarik, 1986; Tesarik, 1988; Braude, 1988].
  • Without wishing to be limited to a particular theory of how IP exerts its effects, it is proposed that embryos exposed to oviduct-derived PGI2 or PGE2 during early development retain the enhanced hatching potential when they reach the uterus. Through oviduct-derived PGI2, the embryos, therefore, actively prepare themselves for implantation while they are in the oviduct. From the perspective of an embryo, oviduct-derived PGI2 is complementary to endometrium-derived PGI2, which mediates endometrial decidualization to ensure receptivity [Lim, 1999].
  • Without wishing to be limited to a particular theory, it is proposed that the enhanced hatching mediated by PGI2 or PGE2 may be due to an increased number of embryonic cells. Different mechanisms are involved in the hatching of mouse embryo in vitro and in vivo. Hatching in vitro involves blastocyst expansion, causing a global zonal thinning prior to zonal rupture, whereas hatching in vivo involves global zonal lysis by uterine or trophectodermal lysins [Montag, 2000]. Therefore, a sufficiently high number of embryonic cells is required to accomplish hatching in vitro. In this respect, our results are consistent with findings that embryos co-cultured with epithelial cells of the oviduct had more cells, less cell death, and improved hatching [Yeung, 1992; Xu, 2000]. In addition, PGI2 may increase production of trypsin-like proteases by the trophectoderm to lyse the zona [Sawada, 1990; Perona, 1986].
  • The low implantation potential of IVF embryos (10-20% per embryo) [CDC, 2001] was attributed in part to sub-optimal culture conditions [De Vos, 2000]. Culture media have been modified to improve embryo development in vitro [Gardner, 1998]. Based on our data, supplementing media with PGI2 and/or PGE2 analog such as Iloprost, PGE1 may provide an improved environment for the developing embryos and increase their implantation potential.
  • In conclusion, PGI2 enhanced the complete hatching of cultured mouse embryos. The stage-specific expression of IP by the mouse embryos coincided with their responsiveness to PGI2. Finally, based on our preliminary data, PGE2 also enhanced the complete hatching of cultured mouse embryos.
  • In further studies we demonstrated that supplementing culture media with PGI2 enhanced the implantation and potential live births of mouse embryos. These results corroborate and extend our initial observation that PGI2 enhanced complete hatching of mouse embryos in culture [Huang, 2003]; they also support our hypothesis that oviduct-derived PGI2 may serve a physiological function [Huang, 2002].
  • Table 1 shows that PGI2 enhanced the implantation potentials of mouse embryos. Eighty-four control embryos and 81 Iloprost-treated embryos were transferred to 12 gestational carriers. Seventy two hours later, more gestation sacs were found in the Iloprost-treated group than the control group (76% versus 42%, relative risk 1.84, 95% confidence interval 1.38-2.43, p<0.0001). At the time of transfer, the developmental stages of the embryos were comparable.
  • Table 2 shows that PGI2 Iloprost enhanced the live birth potentials of mouse embryos. Four hundred six control embryos and 415 Iloprost-treated embryos were transferred to 30 and 31 gestational carriers, respectively. The live birth rates of the control and the experimental groups were 28% and 36%, respectively (p=0.0017, relative risk 1.28, 95% confidence interval: 1.044-1.560). The weights of the pups and the placentae were comparable. Thus, PGI2 increased the potential for live births of mouse embryos without affecting the weight of either the pups or the placentae. In addition, no gross anomalies were noted.
  • As noted above, approximately 70% of experimental and control embryos were at the morula stage at the time of transfer (Table 1), yet the transferred experimental embryos yielded more gestation sacs. These results validate our previous observation that exposure to PGI2 between 8-cell and morula stages was sufficient to enhance the complete hatching of mouse embryos [Huang, 2003]. They are also consistent with oviduct-derived PGI2 serving as an embryotrophic factor, because fertilized mouse eggs develop into morulae inside the oviduct [Snell, 1966].
  • The above-mentioned phenomenon indicates that PGI2 initiates a chain of events culminating in enhanced hatching, implantation, and increased live births. In this regard, PGI2 is reminiscent of platelet-activating factor (PAF) in which short term exposure to PAF increased the implantation of mouse [O'Neill, 1998] and human embryos [O'Neill, 1989]. The molecular and cellular events launched by PGI2 must be critical to hatching. They may include increased embryonic cell number [Montag, 2000], augmented production of trypsin-like proteases by the trophectoderm [Sawada, 1990; Perona, 1986], enhanced blastocoele expansion due to enhanced Na+-K+-ATPase system of the trophectoderm [Biggers, 1988] or yet to be discovered mechanism(s).
  • The implantation rates of control embryos were comparable (42% and 46%) when examined 72 hours or 14 days after transfer, respectively. On the other hand, the implantation rate of experimental embryos examined 14 days after transfer (59%) was significantly less (p=0.038) than that examined 72 hours after transfer (76%). It is plausible that more sacs were completely resorbed in the experimental group due to crowding of implantation sites. Alternatively, Iloprost “rescued” some embryos that would otherwise not have implanted and only a fraction of those “rescued” embryos developed into live pups. Results from this study and abundant PGI2 production by human [Huang, 2002] and mouse [Huang, 2004a] oviducts suggest that PGI2 is one of the embryotrophic factors secreted by the oviducts, and thus supplementing embryo culture media with PGI2 analog is expected to improve IVF outcome.
  • Supplementing IVF culture media with substances that enhance embryo development or implantation has been adopted by most laboratories and media manufacturers with great care. The concerns may include issues regarding reproductive toxicology, teratogenecity, and possibly, epigenetic effects of such manipulation. Lambs from ovine embryos cultured in media supplemented with human serum were heavier and had a longer gestation period [Thompson, 1995; Holm, 1996; Sinclair, 1999]. A congenital disorder involving overgrowth and neoplasia, the Beckwith-Wiedermann syndrome, was associated with assisted reproductive technology [DeBaun, 2002; Maher, 2003].
  • Reproductive toxicology studies on Iloprost [Battenfeld, 1995] showed embryonic and fetal development was not affected in rabbits and monkeys, but digit reduction was observed in rats. The digit anomaly is likely to be due to reduced uteroplacental flow causing hypoxia of the affected structure rather than the inherent teratogenicity of Iloprost, because Iloprost is a vasodilator and the studied animals received Iloprost throughout the gestation period. A similar defect could be induced by vasodilators with different chemical structures such as hydralazine and dihydropyridines. In the current study the exposure to Iloprost took place during the pre-implantation period. Among the 246 live pups examined, there was no digit reduction or other anomalies and the weights of pups and placentae between the two groups were similar. Despite all these, supplementing IVF media with PGI2 analog should be considered an experimental protocol.
  • In these exemplary studies, we demonstrated that PGI2 enhances the potentials of implantation and live births of mouse embryos without affecting the weights of either the fetuses or the placentae, i.e., prostacyclin did not increase the weights of the placenta or the baby. This is an advantage that is separate from the enhancement of implantation and live birth, and indicates that prostacyclin is different from other agents that enhance implantation and live birth. Previously reported agents which enhance implantation and live births also increase the weight of the babies, making those agents unsuitable for human in vitro fertilization (IVF) use. Moreover, because prostacyclin did not affect the weights of the placenta and the babies in the in vivo mouse model, it can be considered for use in human IVF embryos.
  • The results obtained in the foregoing in vivo studies of mouse embryos are considered representative or predictive of similar favorable in vivo results that will be obtained in other mammalian embryos, including human, when those embryos are similarly treated in vitro with prostaglandin or a prostaglandin analog prior to being transferred to the uterus. The current human IVF involves the transfer of 8-cell stage or blastocyst stage embryos to the patient's uterus. In this respect, the human IVF is similar to the design of our experiment involving transferring control or prostacyclin analog-treated embryos to gestational carriers. Our data showed, when transferred to gestational carriers, experimental embryos exposed to prostacyclin analog between 8-cell and morula stages had significantly higher rates of implantation and live births than control embryos (as shown in Tables 1 and 2, respectively).
  • Prostacyclin Agonist (Iloprost) Enhances Human Embryo Development.
  • In the above-described studies, and in WO 2005/026324 (The Board of Regents of the University of Texas System), the disclosure of which is hereby incorporated herein by reference, it is shown that supplementation of Iloprost significantly enhanced mouse embryo development to and beyond blastocyst stage [Huang et al. 2004b]. It was also disclosed that in Iloprost-treated mouse blastocysts transferred to gestational carriers, both the implantation rate and viable pulp rate were significantly increased [Huang et al. 2004c], with no significant difference in body weight between the control and experimental group [Huang et al. 2004c]. The inventors have carried out further studies of Iloprost treatment to enhance the development of human embryos. These human embryo studies were carried out without funding from the United States Government.
  • With the patients' written instructions, 101 frozen zygotes from 14 couples were used for the blastocyst formation study. The zygotes had been frozen during the period of January, 1993 to November, 2003. After overnight culture, only those embryos that had developed to 2-cell stage were used for further study. For each patient, those 2-cell stage embryos were split into a control pool or an experimental pool. All embryos were cultured in Gobal medium (a modified KSOM medium from IVF online) for 5 days with change medium at 2.5 day. The experimental group was supplemented with 0.1 μM of Iloprost (Cayman Chemical, Ann Arbor, Mich.). The control group was supplemented with vehicle (0.000001% DMSO). Chi-Square test were used to compare the number of blastocyst formed. The quality (size, inner cell mass, trophectoderm) of blastocysts was analyzed by non-parametric Wilcoxon Rank Sum Test. The statistical significance was defined at P<0.05. Another 52 donated frozen blastocysts, from 9 couples, were used for short term study. Forty-five thawed blastocysts were recovered. Those blastocysts were immediately cultured with or without Iloprost. The re-expansion of blastocoel was checked 18 hours later. The analysis of outcome was the same as the analysis for the frozen zygotes study. The results of this study are shown in Table 3.
  • Due to the limitation of human embryos available for research, the blastocyst formation rate was at border line (P=0.0506). As more zygotes become available for additional studies, it is expected that a significant increase in the blastocyst formation by Iloprost will be seen. We have reported that the size and quality of trophectoderm correlates with pregnancy [Wun et al. 2001a]. Low quality of inner cell mass correlates with spontaneous abortion [Wun et al. 2001b]. The significant enhancing of these three qualities observed in the present human studies suggests a possible increase in implantation and viable pregnancy rates by Iloprost treatment in human assisted reproductive technology (ART) cases. Although it initially appeared that Iloprost might help the thawed blastocysts, the trend was not significantly different. There are many possible mechanisms that may be responsible for the non-significant difference, such as the trauma from freezing and thawing, the duration of Iloprost treatment, the sensitivity of blastocyst to Iloprost, the required time to show morphological difference, or another mechanism.
  • In summary, this initial study using human embryos showed that Iloprost significantly enhances the quality of blastocysts. The enhancement of the three qualities: size, inner cell mass, and trophectoderm that was observed in the present human studies suggests a possible or probable increase in implantation and viable pregnancy rates by Iloprost treatment in human assisted reproductive technology (ART) cases. Further investigation is expected to clarify the mechanism of action of prostacyclin and its application to human ART.
    TABLE 1
    Treatment
    Control Iloprost p
    Total embryos transferred 84 81
    Degenerated 0 0 n.s.
    Morula 48 49 n.s.
    Early Blastocyst+ 12 8 n.s.
    Late Blastocyst+ 24 22 n.s.
    Hatching Blastocyst 0 2 n.s.
    Number of gestation sacs 35 (42%) 62 (76%) <0.0001*

    n.s. not significant

    *Fisher's exact test, relative risk 1.84 (95% Confidence interval: 1.39-2.43)

    +Early and late blastocysts denote embryos whose blastocoele cavity occupies <49% and ≧50% of the embryo, respectively.
  • TABLE 2
    Treatment
    Control Iloprost p
    Total embryos transferred 406 415
    Number of gestation carriers 30 31
    Total number of live pups 115 (28%) 149 (36%) 0.017*
    Pup weight (gm**) 0.788 ± 0.151 0.772 ± 0.124 n.s.
    Placental weight (gm**) 0.166 ± 0.037 0.166 ± 0.003 n.s.

    n.s. not significant

    *Fisher's exact test, relative risk 1.28 (95% confidence interval: 1.04-1.56).

    **expressed as mean ± S.D.
  • TABLE 3
    Frozen Zygote Frozen blastocyst
    Blastocyst Blastocyst
    formation formate
    rate Size ICM TRO rate Size ICM TRO
    Control 42% 1.42 0.50 0.46 50% 1.45 0.72 0.68
    (11/26) (11/22)
    Iloprost 69% 3.04 1.42 1.31 48% 1.47 0.86 0.78
    (18/26) (11/23)
    Statistical n.s. P < 0.05 P < 0.005 P < 0.005 n.s. n.s. n.s. n.s.
    analysis P = 0.0506

    n.s. not significant

    The score of size is 1, 2, 3, 4, 5, 6. The coding for ICM and TRO is A = 3, B = 2, C = 1. For morula or degenerated embryos coding is 0.
  • REFERENCES
    • 1. Arbab, F., Goldsby, J., Matijevic-Aleksic, N., Huang, G., Ruan, K. H., and Huang, J. C. (2002) Prostacyclin is an autocrine regulator in the contraction of oviductal smooth muscle. Hum Reprod. 17, 3053-9.
    • 2. Battenfeld, R., Schuh, W., and Schobel, C. (1995) Studies on reproductive toxicity of iloprost in rats, rabbits and monkeys. Toxicol Lett. 78, 223-34.
    • 3. Biggers, J. D., Bell, J. E., and Benos, D. J. (1988) Mammalian blastocyst: transport functions in a developing epithelium. Am J. Physiol. 255, C419-32.
    • 4. Braude, P., Bolton, V., and Moore, S. (1988) Human gene expression first occurs between the four- and eight-cell stages of preimplantation development. Nature. 332, 459-61.
    • 5. CDC. (2001). “ART Cycles Using Fresh, Nondonor Eggs or Embryos.” 1999 Assisted Reproductive Technology Success Rates, National Summary and Fertility Clinic Reports, U.S. Department of Health and Human Services Center for Disease Control and Prevention, 35.
    • 6. De Vos, A., and Van Steirteghem, A. (2000) Zona hardening, zona drilling and assisted hatching: new achievements in assisted reproduction. Cells Tissues Organs. 166, 220-7.
    • 7. DeBaun, M. R., Niemitz, E. L., and Feinberg, A. P. (2002) Association of In Vitro Fertilization with Beckwith-Wiedemann Syndrome and Epigenetic Alterations of LIT1 and H19. Am J Hum Genet. 72, 1.
    • 8. Gardner, D. K. (1998) Changes in requirements and utilization of nutrients during mammalian preimplantation embryo development and their significance in embryo culture. Theriogenology. 49, 83-102.
    • 9. Holm, P., Walker, S. K., and Seamark, R. F. (1996) Embryo viability, duration of gestation and birth weight in sheep after transfer of in vitro matured and in vitro fertilized zygotes cultured in vitro or in vivo. J Reprod Fertil. 107, 175-81.
    • 10. Huang, J. C., Arbab, F., Tumbusch, K. J., Goldsby, J. S., Matijevic-Aleksic, N., and Wu, K. K. (2002) Human Fallopian Tubes Express Prostacyclin (PGI) Synthase and Cyclooxygenases and Synthesize Abundant PGI. J Clin Endocrinol Metab. 87, 4361-4368.
    • 11. Huang, J. C., Goldsby, J. S., Arbab, F., Melham, Z., Alecsic, N., and Wu, K. K. (2004a) Oviduct Prostacyclin Functions as a Paracrine Factor to Augment the Development of Embryos. Hum Reprod. 19:2907-12.
    • 12. Huang, J.-C., Wun, W.-S. A., Goldsby, J. S., Wun, I. C., Falconi, S. M., and Wu, K. K. (2003) Prostacyclin enhances embryo hatching but not sperm motility. Hum. Reprod., 18, 2582-2589.
    • 13. Katsuyama, M., Sugimoto, Y., Namba, T., Irie, A., Negishi, M., Narumiya, S., and Ichikawa, A. (1994) Cloning and expression of a cDNA for the human prostacyclin receptor. FEBS Lett. 344, 74-8.
    • 14. Lane, M., and Gardner, D. K. (1997) Differential regulation of mouse embryo development and viability by amino acids. J Reprod Fertil. 109, 153-64.
    • 15. Lim, H., Gupta, R. A., Ma, W. G., Paria, B. C., Moller, D. E., Morrow, J. D., DuBois, R. N., Trzaskos, J. M., and Dey, S. K. (1999) Cyclo-oxygenase-2-derived prostacyclin mediates embryo implantation in the mouse via PPARdelta. Genes Dev. 13, 1561-74.
    • 16. Lim, H., Paria, B. C., Das, S. K., Dinchuk, J. E., Langenbach, R., Trzaskos, J. M., and Dey, S. K. (1997) Multiple female reproductive failures in cyclooxygenase 2-deficient mice. Cell. 91, 197-208.
    • 17. Maher, E. R., Brueton, L. A., Bowdin, S. C., Luharia, A., Cooper, W., Cole, T. R., Macdonald, F., Sampson, J. R., Barratt, C. L., Reik, W., and Hawkins, M. M. (2003) Beckwith-Wiedemann syndrome and assisted reproduction technology (ART). J Med Genet. 40, 62-64.
    • 18. Montag, M., Koll, B., Holmes, P., and van der Ven, H. (2000) Significance of the number of embryonic cells and the state of the zona pellucida for hatching of mouse blastocysts in vitro versus in vivo. Biol Reprod. 62, 1738-44.
    • 19. Murata, T., Ushikubi, F., Matsuoka, T., Hirata, M., Yamasaki, A., Sugimoto, Y., Ichikawa, A., Aze, Y., Tanaka, T., Yoshida, N., Ueno, A., Oh-ishi, S., and Narumiya, S. (1997) Altered pain perception and inflammatory response in mice lacking prostacyclin receptor. Nature. 388, 678-82.
    • 20. O'Neill, C. (1998) Autocrine mediators are required to act on the embryo by the 2-cell stage to promote normal development and survival of mouse preimplantation embryos in vitro. Biol Reprod. 58, 1303-9.
    • 21. O'Neill, C., Ryan, J. P., Collier, M., Saunders, D. M., Ammit, A. J., and Pike, I. L. (1989) Supplementation of in-vitro fertilisation culture medium with platelet activating factor. Lancet. 2, 769-72.
    • 22. Paria, B. C., Huet-Hudson, Y. M., and Dey, S. K. (1993) Blastocyst's state of activity determines the “window” of implantation in the receptive mouse uterus. Proc Natl Acad Sci USA. 90, 10159-62.
    • 23. Perona, R. M., and Wassarman, P. M. (1986) Mouse blastocysts hatch in vitro by using a trypsin-like proteinase associated with cells of mural trophectoderm. Dev Biol. 114, 42-52.
    • 24. Piekos, M. W., Frasor, J., Mack, S., Binor, Z., Soltes, B., Molo, M. W., Radwanska, E., and Rawlins, R. G. (1995) Evaluation of co-culture and alternative culture systems for promoting in-vitro development of mouse embryos. Hum Reprod. 10, 1486-91.
    • 25. Sawada, H., Yamazaki, K., and Hoshi, M. (1990) Trypsin-like hatching protease from mouse embryos: evidence for the presence in culture medium and its enzymatic properties. J Exp Zool. 254, 83-7.
    • 26. Sinclair, K. D., McEvoy, T. G., Maxfield, E. K., Maltin, C. A., Young, L. E., Wilmut, I., Broadbent, P. J., and Robinson, J. J. (1999) Aberrant fetal growth and development after in vitro culture of sheep zygotes. J Reprod Fertil. 116, 177-86.
    • 27. Snell, G. D., and Stevens, L. C. (1966). “Early embryology.” Biology of the Laboratory Mouse by the staff of the Jackson Laboratory, E. L. Green, ed., Blakiston Division, McGraw-Hill Book Company, New York, 205-245.
    • 28. Tesarik, J., Kopecny, V., Plachot, M., and Mandelbaum, J. (1986) Activation of nucleolar and extranucleolar RNA synthesis and changes in the ribosomal content of human embryos developing in vitro. J Reprod Fertil. 78, 463-70.
    • 29. Tesarik, J., Kopecny, V., Plachot, M., and Mandelbaum, J. (1988) Early morphological signs of embryonic genome expression in human preimplantation development as revealed by quantitative electron microscopy. Dev Biol. 128, 15-20.
    • 30. Thompson, J. G., Gardner, D. K., Pugh, P. A., McMillan, W. H., and Tervit, H. R. (1995) Lamb birth weight is affected by culture system utilized during in vitro pre-elongation development of ovine embryos. Biol Reprod. 53, 1385-91.
    • 31. Tsai, A. L., Hsu, M. J., Vijjeswarapu, H., and Wu, K. K. (1989) Solubilization of prostacyclin membrane receptors from human platelets. J Biol. Chem. 264, 61-7.
    • 32. Xu, J., Cheung, T. M., Chan, S. T., Ho, P. C., and Yeung, W. S. (2000) Human oviductal cells reduce the incidence of apoptosis in cocultured mouse embryos. Fertil Steril. 74, 1215-9.
    • 33. Xu, J. S., Cheung, T. M., Chan, S. T., Ho, P. C., and Yeung, W. S. (2001) Temporal effect of human oviductal cell and its derived embryotrophic factors on mouse embryo development. Biol Reprod. 65, 1481-8.
    • 34. Yeung, W. S., Ho, P. C., Lau, E. Y., and Chan, S. T. (1992) Improved development of human embryos in vitro by a human oviductal cell co-culture system. Hum Reprod. 7, 1144-9.
    • 35. Huang, J.-C., Goldsby, J. S. and Wun, W.-S. A. (2004b) Prostayclin enhances the implantation and live birth potentials of mouse embryos. Hum Reprod 19:1856-1860.
    • 36. Huang, J. C., Win, W. S., Goldsby, J. S., Matijevic-Aleksic, N., Wu, K. K. (2004c) Hum Reprod 19:2900-2906.
    • 37. Wun, W. A., Grunert, G. M., Dunn, R. C., Valdes, C. T., Schenk, L. M., Mangal, R. K. (2001a) Fert. Steril. 76(Suppl. 1):S143-S144.
    • 38. Wun, W-S. A., Grunert, G. M., Dunn, R., Valdes, C., Schenk, L., Mangal, R. (2001b) Fert. Steril. 78(Suppl. 1):S137-S138.
  • Without further elaboration, it is believed that one skilled in the art can, using the description herein, utilize the present invention to its fullest extent. The foregoing embodiments are to be construed as illustrative, and not as constraining the remainder of the disclosure in any way whatsoever. While the preferred embodiments of the invention have been shown and described, modifications thereof can be made by one skilled in the art without departing from the spirit and teachings of the invention. The embodiments described herein are exemplary only, and are not intended to be limiting. Many variations and modifications of the invention disclosed herein are possible and are within the scope of the invention. Any discussion of references in the Background of the Invention is not an admission that it is prior art to the present invention, especially any reference that may have a publication date after the priority date of this application. The disclosures of all patents, patent applications and publications cited herein are hereby incorporated herein by reference, to the extent that they provide exemplary, procedural or other details supplementary to those set forth herein.

Claims (11)

1. An in vitro method of enhancing the development of a mammalian embryo to at least the blastocyst stage, comprising:
obtaining an in vitro fertilization embryo comprising at least two cells;
growing said embryo in a culture medium supplemented with an amount of a prostaglandin or a prostaglandin analog effective for enhancing the quality of a resulting blastocyst.
2. The method of claim 1 wherein said enhancing comprises increasing blastocyst size.
3. The method of claim 1 wherein said enhancing comprises increasing inner cell mass of said blastocyst.
4. The method of claim 1 wherein said enhancing comprises increasing trophectoderm size of said blastocyst.
5. The method of claim 1 wherein said enhancing comprises increasing at least two qualities chosen from the group consisting of blastocyst size, inner cell mass and trophectoderm size.
6. The method of claim 1 wherein said prostaglandin is prostacyclin (PGI2) or prostaglandin E2 (PGE2).
7. The method of claim 1 wherein said prostaglandin analog comprises a prostacyclin agonist.
8. The method of claim 7 wherein said agonist comprises Iloprost.
9. The method of claim 7 wherein said prostaglandin analog comprises prostaglandin E1 (PGE1).
10. A method of increasing the in vivo implantation potential of an in vitro fertilization embryo comprising:
culturing a mammalian zygote in vitro in a culture medium supplemented with an amount of a prostaglandin or a prostaglandin analog effective to promote an increase in quality of resulting blastocysts, whereby the in vivo implantation potential of a resulting embryo is enhanced.
11. The method of claim 10, wherein said culturing of a mammalian zygote in a culture medium supplemented with said prostaglandin or prostaglandin analog further enhances establishment of a viable pregnancy when the resulting embryo is implanted in a uterus.
US11/370,152 2003-09-08 2006-03-07 Enhancement of mammalian embryo development Expired - Fee Related US7709259B2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP04783425A EP1670902A4 (en) 2003-09-08 2004-09-08 METHOD AND COMPOSITION FOR ENHANCING i IN VITRO /i EMBRYO DEVELOPMENT BY SUPPLEMENTING CULTURE MEDIUM WITH PROSTAGLANDIN OR A PROSTAGLANDIN ANALOG
PCT/US2004/029167 WO2005026324A2 (en) 2003-09-08 2004-09-08 Method and composition for enhancing in vitro embryo development by supplementing culture medium with prostaglandin or a prostaglandin analog
US11/370,152 US7709259B2 (en) 2003-09-08 2006-03-07 Enhancement of mammalian embryo development

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US50116603P 2003-09-08 2003-09-08
PCT/US2004/029167 WO2005026324A2 (en) 2003-09-08 2004-09-08 Method and composition for enhancing in vitro embryo development by supplementing culture medium with prostaglandin or a prostaglandin analog
US70829105P 2005-08-15 2005-08-15
US11/370,152 US7709259B2 (en) 2003-09-08 2006-03-07 Enhancement of mammalian embryo development

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/029167 Continuation-In-Part WO2005026324A2 (en) 2003-09-08 2004-09-08 Method and composition for enhancing in vitro embryo development by supplementing culture medium with prostaglandin or a prostaglandin analog

Publications (3)

Publication Number Publication Date
US20060160213A1 US20060160213A1 (en) 2006-07-20
US20090011496A9 true US20090011496A9 (en) 2009-01-08
US7709259B2 US7709259B2 (en) 2010-05-04

Family

ID=37261816

Family Applications (1)

Application Number Title Priority Date Filing Date
US11/370,152 Expired - Fee Related US7709259B2 (en) 2003-09-08 2006-03-07 Enhancement of mammalian embryo development

Country Status (3)

Country Link
US (1) US7709259B2 (en)
EP (1) EP1670902A4 (en)
WO (1) WO2005026324A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160118494A1 (en) * 2013-05-17 2016-04-28 Robert Bosch Gmbh Mos field-effect transistor and method for the production thereof
US20160190256A1 (en) * 2013-10-02 2016-06-30 Infineon Technologies Ag Semiconductor Device Including a Transistor with a Gate Dielectric Having a Variable Thickness

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1933618A4 (en) * 2005-10-12 2010-01-13 Univ Texas Peroxisome proliferator-activator receptor delta (ppar delta) and the development of preimplantation embryos
WO2010010201A1 (en) 2008-07-22 2010-01-28 Equipo Ivi Investigacion Sl Gene expression profile as a marker of endometrial receptivity
EP4012023B1 (en) 2014-06-17 2024-05-15 Asherman Therapy, S.L. Stem cell therapy in endometrial pathologies
CN104166001A (en) * 2014-07-24 2014-11-26 中国农业科学院北京畜牧兽医研究所 Kit for detecting mammal blastula inner cell mass (ICM)/trophectoderm (TE) ratio and cell apoptosis
WO2017125918A1 (en) * 2016-01-18 2017-07-27 Amorphical Ltd. Stabilized amorphous calcium carbonate for treatment of neurological, muscular and infertility diseases or conditions

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5096822A (en) * 1990-07-26 1992-03-17 W. R. Grace & Co.- Conn. Bovine embryo medium
US6100249A (en) * 1992-12-30 2000-08-08 Applied Research Systems Ars Holdings Nv Use of aspirin for the manufacture of a medicament for implanting an embryo into the uterine endometrium
US20020088019A1 (en) * 1997-09-02 2002-07-04 Oron Yacoby-Zeevi Methods of and pharmaceutical compositions for improving implantation of embryos

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003517276A (en) 1998-11-30 2003-05-27 アイ・ブイ・エフ・サイエンシイズ・コロラド・インコーポレーテツド Systems and continuous cultures for in vitro fertilization

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5096822A (en) * 1990-07-26 1992-03-17 W. R. Grace & Co.- Conn. Bovine embryo medium
US6100249A (en) * 1992-12-30 2000-08-08 Applied Research Systems Ars Holdings Nv Use of aspirin for the manufacture of a medicament for implanting an embryo into the uterine endometrium
US20020088019A1 (en) * 1997-09-02 2002-07-04 Oron Yacoby-Zeevi Methods of and pharmaceutical compositions for improving implantation of embryos

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160118494A1 (en) * 2013-05-17 2016-04-28 Robert Bosch Gmbh Mos field-effect transistor and method for the production thereof
US20160190256A1 (en) * 2013-10-02 2016-06-30 Infineon Technologies Ag Semiconductor Device Including a Transistor with a Gate Dielectric Having a Variable Thickness

Also Published As

Publication number Publication date
US7709259B2 (en) 2010-05-04
EP1670902A4 (en) 2008-05-07
WO2005026324A2 (en) 2005-03-24
WO2005026324A3 (en) 2005-07-14
US20060160213A1 (en) 2006-07-20
EP1670902A2 (en) 2006-06-21

Similar Documents

Publication Publication Date Title
Eppig et al. Development in vitro of mouse oocytes from primordial follicles
Fry et al. Human leukemia inhibitory factor improves the viability of cultured ovine embryos
US7709259B2 (en) Enhancement of mammalian embryo development
Edwards Human implantation: the last barrier in assisted reproduction technologies?
KR20180079302A (en) Culture method for differentiating gonadal cells into functionally mature oocyte cells
JP2003533976A (en) Clonal propagation of primate offspring by embryo division
Lim et al. Roles of growth factors in the development of bovine embryos fertilized in vitro and cultured singly in a defined medium
KR102306054B1 (en) Composition and Method for enhancing developmental rate of embryos using melatonin
Rath et al. In vitro development to blastocysts of early porcine embryos produced in vivo or in vitro
Feng et al. Fertilization and early embryology: Effect of different co-culture systems in early human embryo development
Prichard et al. In-vitro co-culture of early stage caprine embryos with oviduct and uterine epithelial cells
Quinn Use of coculture with cumulus cells in insemination medium in human in vitro fertilization (IVF)
Seita et al. Full-term development of rats from oocytes fertilized in vitro using cryopreserved ejaculated sperm
JP3639898B2 (en) Culture medium for in vitro culture of porcine in vitro produced embryo and method for in vitro culture of porcine in vitro produced embryo using the culture medium
JP2003512833A (en) METHODS AND COMPOSITIONS FOR ENHANCED DEVELOPMENTAL CAPABILITIES OF OOCYTES AND ZYGOTS
Lopata History of the Egg in Embryology
Thibault Recent data on the development of cloned embryos derived from reconstructed eggs with adult cells
JP2007504807A (en) Methods and compositions for enhancing in vitro embryogenesis by supplementing culture media with prostaglandins or prostaglandin analogs
EP3598895A1 (en) An in vitro method for freezing mammalian embryos
Edwards et al. Future development in IVF
WO2005045007A1 (en) Method and system for storing stem cell in frozen state
Yamatoya et al. The fluid factor OVGP1 provides a significant oviductal microenvironment for the reproductive process in golden hamster
JPH08289779A (en) Serum-free culture medium and production of in vitro fertilized egg
CN116445396A (en) New application of WNT2 in improving development efficiency and quality of in vitro fertilized embryo
Yamatoya et al. The fluid factor OVGP1 provides a significant oviductal microenvironment for the

Legal Events

Date Code Title Description
AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF TEXAS HLTH SCI CTR HOUSTON;REEL/FRAME:022466/0287

Effective date: 20090309

AS Assignment

Owner name: BOARD OF REGENTS OF THE UNIVERSITY OF TEXAS SYSTEM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HUANG, JAOU-CHEN;GOLDSBY, JENNIFER S.;WUN, WAN-SONG A.;REEL/FRAME:023824/0310;SIGNING DATES FROM 20100112 TO 20100113

Owner name: BOARD OF REGENTS OF THE UNIVERSITY OF TEXAS SYSTEM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HUANG, JAOU-CHEN;GOLDSBY, JENNIFER S.;WUN, WAN-SONG A.;SIGNING DATES FROM 20100112 TO 20100113;REEL/FRAME:023824/0310

FPAY Fee payment

Year of fee payment: 4

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.)

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.)

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20180504