Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
CRISPR J ; 7(1): 12-28, 2024 02.
Article in English | MEDLINE | ID: mdl-38353617

ABSTRACT

Disease resistance genes in livestock provide health benefits to animals and opportunities for farmers to meet the growing demand for affordable, high-quality protein. Previously, researchers used gene editing to modify the porcine CD163 gene and demonstrated resistance to a harmful virus that causes porcine reproductive and respiratory syndrome (PRRS). To maximize potential benefits, this disease resistance trait needs to be present in commercially relevant breeding populations for multiplication and distribution of pigs. Toward this goal, a first-of-its-kind, scaled gene editing program was established to introduce a single modified CD163 allele into four genetically diverse, elite porcine lines. This effort produced healthy pigs that resisted PRRS virus infection as determined by macrophage and animal challenges. This founder population will be used for additional disease and trait testing, multiplication, and commercial distribution upon regulatory approval. Applying CRISPR-Cas to eliminate a viral disease represents a major step toward improving animal health.


Subject(s)
Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Animals , Swine , Porcine respiratory and reproductive syndrome virus/genetics , Porcine Reproductive and Respiratory Syndrome/genetics , CRISPR-Cas Systems/genetics , Disease Resistance/genetics , Gene Editing , Livestock
2.
Physiol Genomics ; 47(4): 102-12, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25586978

ABSTRACT

Oocytes isolated from female rhesus monkeys following standard ovarian stimulation protocols during the summer months displayed a reduced capacity to mature compared with stimulation during the normal breeding season. Because the gene expression profiles of oocyte-associated cumulus cells and mural granulosa cells (CCs and GCs) are indicative of altered oocyte quality and can provide insight into intrafollicular processes that may be disrupted during oogenesis, we performed array-based transcriptome comparisons of CCs and GCs from summer and normal breeding season stimulation cycles. Summer CCs and GCs both display deficiencies in expression of mRNAs related to cell proliferation, angiogenesis, and endocrine signaling, as well as reduced expression of glycogen phosphorylase. Additionally, CCs display deficiencies in expression of mRNAs related to stress response. These results provide the first insight into the specific molecular pathways and processes that are disrupted in the follicles of rhesus macaque females during the summer season. Some of the changes seen in summer GCs and CCs have been reported in humans and in other model mammalian species. This suggests that the seasonal effects seen in the rhesus monkey may help us to understand better the mechanisms that contribute to reduced oocyte quality and fertility in humans.


Subject(s)
Macaca mulatta/physiology , Oocytes/growth & development , Ovarian Follicle/physiology , Ovary/physiology , Animals , Female , Gene Expression Profiling , Male , Oocytes/cytology , Ovary/cytology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Seasons
3.
Fertil Steril ; 103(2): 560-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25492684

ABSTRACT

OBJECTIVE: To determine if binge ethanol consumption before ovulation affects oocyte quality, gene expression, and subsequent embryo development. DESIGN: Binge levels of ethanol were given twice weekly for 6 months, followed by a standard in vitro fertilization cycle and subsequent natural mating. SETTING: National primate research center. ANIMAL(S): Adult female rhesus monkeys. INTERVENTION(S): Binge levels of ethanol, given twice weekly for 6 months before a standard in vitro fertilization cycle with or without embryo culture. With in vivo development, ethanol treatment continued until pregnancy was identified. MAIN OUTCOME MEASURE(S): Oocyte and cumulus/granulosa cell gene expression, embryo development to blastocyst, and pregnancy rate. RESULT(S): Embryo development in vitro was reduced; changes were found in oocyte and cumulus cell gene expression; and spontaneous abortion during very early gestation increased. CONCLUSION(S): This study provides evidence that binge drinking can affect the developmental potential of oocytes even after alcohol consumption has ceased.


Subject(s)
Binge Drinking/complications , Binge Drinking/pathology , Ethanol/toxicity , Health Status , Models, Animal , Animals , Cohort Effect , Ethanol/administration & dosage , Female , Humans , Macaca mulatta , Oocytes/drug effects , Oocytes/pathology , Pregnancy , Pregnancy Rate/trends
4.
Adv Exp Med Biol ; 759: 89-110, 2014.
Article in English | MEDLINE | ID: mdl-25030761

ABSTRACT

Post-translational modifications of cellular proteins by ubiquitin and ubiquitin-like protein modifiers are important regulatory events involved in diverse aspects of gamete and embryo physiology including oocyte maturation, fertilization and development of embryos to term. Deubiquitinating enzymes (DUBs) regulate proteolysis by reversing ubiquitination, which targets proteins to the 26S proteasome. The ubiquitin C-terminal hydrolases (UCHs) comprise are DUBs that play a role in the removal of multi-ubiquitin chains. We review here the roles of UCHs in oocytes maturation, fertilization and development in mouse, bovine, porcine and rhesus monkeys. Oocyte UCHs contributes to fertilization and embryogenesis by regulating the physiology of the oocyte and blastomere cortex as well as oocyte spindle. Lack of UCHs in embryos reduces fertilization, while mutant embryos fail to undergo compaction and blastocyst formation. In addition to advancing our understanding of reproductive process, research on the role of deubiquitinating enzymes will allow us to better understand and treat human infertility, and to optimize reproductive performance in agriculturally important livestock species.


Subject(s)
Embryonic Development/physiology , Fertilization/physiology , Oocytes/physiology , Ubiquitin Thiolesterase/physiology , Animals , Humans , Proteasome Endopeptidase Complex/physiology , Ubiquitination
5.
Endocrinology ; 155(7): 2688-95, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24731100

ABSTRACT

The consumption of refined sugars continues to pose a significant health risk. However, nearly nothing is known about the effects of sugar intake by healthy women on the oocyte or embryo. Using rhesus monkeys, we show that low-dose sucrose intake over a 6-month period has an impact on the oocyte with subsequent effects on the early embryo. The ability of oocytes to resume meiosis was significantly impaired, although the differentiation of the somatic component of the ovarian follicle into progesterone-producing cells was not altered. Although the small subset of oocytes that did mature were able to be fertilized in vitro and develop into preimplantation blastocysts, there were >1100 changes in blastocyst gene expression. Because sucrose treatment ended before fertilization, the effects of sugar intake by healthy primates are concluded to be epigenetic modifications to the immature oocyte that are manifest in the preimplantation embryo.


Subject(s)
Dietary Sucrose/administration & dosage , Embryonic Development/drug effects , Oocytes/drug effects , Transcriptome/drug effects , Animals , Blastocyst/drug effects , Blastocyst/metabolism , Cells, Cultured , Female , Fertilization in Vitro , Humans , Macaca mulatta , Meiosis/drug effects , Meiosis/genetics , Oligonucleotide Array Sequence Analysis , Oocytes/growth & development , Oocytes/metabolism , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome/genetics
6.
J Cell Physiol ; 227(4): 1592-603, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21678411

ABSTRACT

Post-translational protein modification by ubiquitination, a signal for lysosomal or proteasomal proteolysis, can be regulated and reversed by deubiquitinating enzymes (DUBs). This study examined the roles of UCHL1 and UCHL3, two members of ubiquitin C-terminal hydrolase (UCH) family of DUBs, in murine fertilization and preimplantation development. Before fertilization, these proteins were associated with the oocyte cortex (UCHL1) and meiotic spindle (UCHL3). Intracytoplasmic injection of the general UCH-family inhibitor ubiquitin-aldehyde (UBAL) or antibodies against UCHL3 into mature metaphase II oocytes blocked fertilization by reducing sperm penetration of the zona pellucida and incorporation into the ooplasm, suggesting a role for cortical UCHL1 in sperm incorporation. Both UBAL and antibodies against UCHL1 injected at the onset of oocyte maturation (germinal vesicle stage) reduced the fertilizing ability of oocytes. The subfertile Uchl1(gad-/-) mutant mice showed an intriguing pattern of switched UCH localization, with UCHL3 replacing UCHL1 in the oocyte cortex. While fertilization defects were not observed, the embryos from homozygous Uchl1(gad-/-) mutant females failed to undergo morula compaction and did not form blastocysts in vivo, indicating a maternal effect related to UCHL1 deficiency. We conclude that the activity of oocyte UCHs contributes to fertilization and embryogenesis by regulating the physiology of the oocyte and blastomere cortex.


Subject(s)
Embryonic Development/physiology , Fertilization/physiology , Ubiquitin Thiolesterase/physiology , Animals , Embryonic Development/genetics , Female , Fertilization/drug effects , Fertilization/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Oocytes/drug effects , Oocytes/enzymology , Oogenesis/physiology , Pregnancy , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/deficiency , Ubiquitin Thiolesterase/genetics , Ubiquitins/pharmacology
7.
J Cell Physiol ; 227(5): 2022-9, 2012 May.
Article in English | MEDLINE | ID: mdl-21751213

ABSTRACT

Ubiquitin C-terminal hydrolases (UCHs) comprise a family of deubiquitinating enzymes that play a role in the removal of multi-ubiquitin chains from proteins that are posttranslationally modified by ubiquitination to be targeted for proteolysis by the 26S proteasome. The UCH-enzymes also generate free monomeric ubiquitin from precursor multi-ubiquitin chains and, in some instances, may rescue ubiquitinated proteins from degradation. This study examined the roles of two oocyte-expressed UCHs, UCHL1, and UCHL3 in murine and rhesus monkey oocyte maturation. The Uchl1 and Uchl3 mRNAs were highly expressed in GV and MII oocytes, and were associated with the oocyte cortex (UCHL1) and meiotic spindle (UCHL3). Microinjection of the UCH-family enzyme inhibitor, ubiquitin-aldehyde (UBAL) to GV oocytes prevented oocyte meiotic progression beyond metaphase I in a majority of treated oocytes and caused spindle and first polar body anomalies. Injection of antibodies against UCHL3 disrupted oocyte maturation and caused meiotic anomalies, including abnormally long meiotic spindles. A selective, cell permeant inhibitor of UCHL3, 4, 5, 6, 7-tetrachloroidan-1, 3-dione also caused meiotic defects and chromosome misalignment. Cortical granule localization in the oocyte cortex was disrupted by UBAL injected after oocyte maturation. We conclude that the activity of oocyte UCHs contributes to oocyte maturation by regulating the oocyte cortex and meiotic spindle.


Subject(s)
Oocytes/physiology , Ubiquitin Thiolesterase/metabolism , Ubiquitins/metabolism , Animals , Female , Humans , Macaca mulatta , Meiosis/physiology , Mice , Microtubules/metabolism , Oocytes/cytology , Spindle Apparatus/metabolism , Ubiquitin Thiolesterase/genetics
8.
Fertil Steril ; 96(1): 203-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21645893

ABSTRACT

OBJECTIVE: To determine whether preimplantation embryos are targets for relaxin secreted from the corpus luteum of the menstrual cycle. DESIGN: Rhesus monkey oocytes obtained from females undergoing controlled ovarian hyperstimulation were inseminated, and the resulting embryos were cultured in medium with or without recombinant human relaxin (20 ng/mL) for 8 days. SETTING: Research laboratory. ANIMAL(S): Rhesus monkey. INTERVENTION(S): Controlled ovarian stimulation to obtain oocytes for in vitro-produced embryos that were cultured with or without human recombinant relaxin. MAIN OUTCOME MEASURE(S): Rate of blastocyst development, percentage of blastocysts, and inner cell mass/trophectoderm cell ratio were measured on day 8 of culture. The presence of relaxin receptor (RXFP1) messenger RNA in eight-cell embryos was observed by array hybridization. RESULT(S): RXFP1 receptor expression was localized to the inner cell mass of blastocysts, as shown by immunohistochemistry. The percentage of embryos that developed to blastocyst and the inner cell mass/trophectoderm cell ratio was unchanged with relaxin supplementation; however, the relaxin-treated embryos developed into blastocysts significantly sooner than untreated embryos. CONCLUSION(S): These results are the first evidence that the preimplantation primate embryo is a target for relaxin and that the addition of relaxin to in vitro culture medium enhances rhesus monkey embryo development.


Subject(s)
Blastocyst/drug effects , Blastocyst/metabolism , Drug Delivery Systems/methods , Relaxin/administration & dosage , Relaxin/metabolism , Animals , Cricetinae , Embryo Culture Techniques/methods , Female , Humans , Macaca mulatta , Pregnancy , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Species Specificity
9.
Gene Expr Patterns ; 11(3-4): 285-98, 2011.
Article in English | MEDLINE | ID: mdl-21329766

ABSTRACT

Two essential aspects of mammalian development are the progressive specialization of cells toward different lineages, and the maintenance of progenitor cells that will give rise to the differentiated components of each tissue and also contribute new cells as older cells die or become injured. The transition from totipotentiality to pluripotentiality, to multipotentiality, to monopotentiality, and then to differentiation is a continuous process during development. The ontological relationship between these different stages is not well understood. We report for the first time an ontological survey of expression of 45 putative "stemness" and "pluripotency" genes in rhesus monkey oocytes and preimplantation stage embryos, and comparison to the expression in the inner cell mass, trophoblast stem cells, and a rhesus monkey (ORMES6) embryonic stem cell line. Our results reveal that some of these genes are not highly expressed in all totipotent or pluripotent cell types. Some are predominantly maternal mRNAs present in oocytes and embryos before transcriptional activation, and diminishing before the blastocyst stage. Others are well expressed in morulae or early blastocysts, but are poorly expressed in later blastocysts or ICMs. Also, some of the genes employed to induce pluripotent stem cells from somatic cells (iPS genes) appear unlikely to play major roles as stemness or pluripotency genes in normal embryos.


Subject(s)
Blastocyst Inner Cell Mass/metabolism , Gene Expression Regulation, Developmental , Macaca mulatta/genetics , Oocytes/metabolism , Pluripotent Stem Cells/metabolism , Animals , Antigens, Differentiation/metabolism , Cells, Cultured , Ectoderm/metabolism , Embryonic Stem Cells/metabolism , Endoderm/metabolism , Female , Gene Expression Profiling , Macaca mulatta/embryology , Mesoderm/metabolism , RNA, Messenger/metabolism , Totipotent Stem Cells/metabolism , Transcription, Genetic , Trophoblasts/metabolism
10.
Am J Physiol Cell Physiol ; 298(5): C1235-44, 2010 May.
Article in English | MEDLINE | ID: mdl-20107036

ABSTRACT

Blastomere cytofragmentation in mammalian embryos poses a significant problem in applied and clinical embryology. Mouse two-cell-stage embryos display strain-dependent differences in the rate of cytofragmentation, with a high rate observed in C3H/HeJ embryos and a lower rate observed in C57BL/6 embryos. The maternally inherited genome exerts the strongest effect on the process, with lesser effects mediated by the paternally inherited genome and the ooplasm. The effect of the maternal genome is transcription dependent and independent of the mitochondrial strain of origin. To identify molecular mechanisms that underlie cytofragmentation, we evaluated transcriptional activities of embryos possessing maternal pronuclei (mPN) of different origins. The mPN from C57BL/6 and C3H/HeJ strains directed specific transcription at the two-cell stage of mRNAs corresponding to 935 and 864 Affymetrix probe set IDs, respectively. Comparing transcriptomes of two-cell-stage embryos with different mPN revealed 64 transcribed genes with differential expression (1.4-fold or greater). Some of these genes occupy molecular pathways that may regulate cytofragmentation via a combination of effects related to apoptosis and effects on the cytoskeleton. These results implicate specific molecular mechanisms that may regulate cytofragmentation in early mammalian embryos. The most striking effect of mPN strain of origin on gene expression was on adenylate cyclase 2 (Adcy2). Treatment with dibutyryl cAMP (dbcAMP) elicits a high rate and severe form of cytofragmentation, and the effective dbcAMP concentration varies with maternal genotype. An activator of exchange proteins directly activated by cAMP (EPACs, or RAPGEF 3 and 4) 8-pCPT-2'-O-methyl-cAMP, elicits a high level of fragmentation while the PKA-specific activator N6-benzoyl-cAMP does not. Inhibition of A kinase anchor protein activities with st-Ht31 induces fragmentation. Inhibition of phosphatidylinositol 3-kinase signaling also induces fragmentation. These results reveal novel mechanisms by which maternal genotype affects cytofragmentation, including a system of opposing signaling pathways that most likely operate by controlling cytoskeletal function.


Subject(s)
Blastomeres/metabolism , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental/physiology , Genome , Transcription, Genetic/physiology , Animals , Blastomeres/cytology , Embryonic Development/physiology , Genotype , Mice , Mice, Inbred C3H , Mice, Inbred C57BL
11.
Biol Reprod ; 81(6): 1139-46, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19641179

ABSTRACT

There is a vital need to identify factors that enhance human and nonhuman primate in vitro embryo culture and outcome, and to identify the factors that facilitate that objective. Granulosa and cumulus cells were obtained from rhesus monkeys that had either been FSH-primed (in vitro maturation [IVM]) or FSH and hCG-primed (in vivo maturation [VVM]) and compared for the expression of mRNAs encoding follistatin (FST), inhibin, and activin receptors. The FST mRNA displayed marginally decreased expression (P = 0.05) in association with IVM in the granulosa cells. The ACVR1B mRNA was more highly expressed in cumulus cells with IVM compared with VVM. Cumulus-oocyte complexes from FSH-primed monkeys exposed to exogenous FST during the 24-h IVM period exhibited no differences in the percentage of oocytes maturing to the metaphase II stage of meiosis compared to controls. However, embryos from these oocytes had significantly decreased development to the blastocyst stage. The effect of FST on early embryo culture was determined by exposing fertilized VVM oocytes to exogenous FST from 12 to 60 h postinsemination. FST significantly improved time to first cleavage and embryo development to the blastocyst stage compared with controls. The differential effects of exogenous FST on embryo development, when administered before and after oocyte maturation, may depend on the endogenous concentration in cumulus cells and oocytes. These results reveal evolutionary conservation of a positive effect of FST on embryogenesis that may be broadly applicable to enhance in vitro embryogenesis, with potential application to human clinical outcome and livestock and conservation biology.


Subject(s)
Blastocyst/drug effects , Embryonic Development/drug effects , Follistatin/metabolism , Follistatin/pharmacology , Oocytes/drug effects , Oogenesis/drug effects , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Animals , Blastocyst/metabolism , Cell Count , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Culture Media , Cumulus Cells/drug effects , Cumulus Cells/metabolism , Embryo Culture Techniques , Embryonic Development/physiology , Female , Fertilization in Vitro , Follicle Stimulating Hormone/pharmacology , Follistatin/genetics , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Inhibins/genetics , Inhibins/metabolism , Macaca mulatta , Oocytes/metabolism , Oogenesis/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Mol Reprod Dev ; 76(3): 255-69, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18646051

ABSTRACT

MicroRNAs (miRNAs) are a class of small RNAs that silence gene expression. In animal cells, miRNAs bind to the 3' untranslated regions of specific mRNAs and inhibit their translation. The correct regulation of mRNA expression by miRNAs is believed to be important for oocyte maturation, early development and implantation. We examined the expression of 25 mRNAs involved in the microRNA processing pathway in a nonhuman primate oocyte and embryo model. We observed that mRNAs related to miRNA splicing are downregulated during oocyte maturation while those related to miRNA processing are upregulated, indicating that there may exist a temporal difference in their activities related to transcriptional activity in germinal vesicle stage oocytes. We also observed that the vast majority of mRNAs examined were insensitive to alpha-amanitin at the 8-16 cell stage. The expression data did not reveal a major impact of embryo culture, and hormonal stimulation protocol affected only a small number of mRNAs, suggesting that the components of the pathway may be accumulated in the oocyte during oogenesis and resistant to exogenous insults. In comparison to published mouse array data, we observed species differences and similarities in the temporal expression patterns of some genes, suggesting that miRNA processing may be regulated differently. These data extend our understanding of the potential roles of miRNA during primate embryogenesis.


Subject(s)
Embryo, Mammalian/metabolism , Macaca mulatta/genetics , MicroRNAs/metabolism , Oocytes/metabolism , Alpha-Amanitin/pharmacology , Animals , Embryonic Development/genetics , Female , Follicle Stimulating Hormone , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Mice , MicroRNAs/genetics , Nucleic Acid Synthesis Inhibitors/pharmacology , Oligonucleotide Array Sequence Analysis , RNA Editing/drug effects , RNA Editing/genetics , RNA Splicing/drug effects , RNA Splicing/genetics , RNA Transport/genetics , SMN Complex Proteins/genetics , SMN Complex Proteins/metabolism , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Biol Reprod ; 80(3): 464-72, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19073997

ABSTRACT

New techniques to boost male and female fertility are being pioneered at a rapid pace in fertility clinics to increase the efficiency of assisted reproduction methods in couples in which natural conception has not been achieved. This study investigates the possible epigenetic effects of ooplasm manipulation methods on postnatal growth and development using a mouse genetic model, with particular emphasis on the possible effects of intergenotype manipulations. We performed interstrain and control intrastrain maternal pronuclear transfers, metaphase-II spindle transfers, and ooplasm transfer between C57BL/6 and DBA/2 mice, and found no major, long-term growth defects or epigenetic abnormalities, in either males or females, associated with intergenotype transfers. Ooplasm transfer itself was associated with reduced viability, and additional subtle effects of ooplasm strain of origin were observed. Both inter- and intrastrain ooplasm transfer were associated with subtle, transient effects on growth early in life. We also performed inter- and intrastrain germinal vesicle transfers (GVTs). Interstrain GVT females, but not males, had significantly lower body weights at birth and thereafter compared with the intrastrain GVT and non-GVT controls. No GVT-associated changes were observed in DNA methylation of the Mup1, Rasgrf1, H19, Snrpn, or Peg3 genes, nor any difference in expression of the imprinted Rasgrf1, Igf2r, or Mest genes. These results indicate that some ooplasm manipulation procedures may exert subtle effects on growth early in life, while intergenotype GVT can result in significant growth deficiencies after birth.


Subject(s)
Animals, Newborn/growth & development , Cytoplasm/transplantation , DNA Methylation/physiology , Oocytes/cytology , Zygote Intrafallopian Transfer , Animals , Embryo Culture Techniques , Epigenesis, Genetic , Female , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Models, Animal , Proteins/metabolism , Spindle Apparatus/transplantation , ras-GRF1/metabolism
14.
Int Rev Cell Mol Biol ; 268: 223-90, 2008.
Article in English | MEDLINE | ID: mdl-18703408

ABSTRACT

The oocyte is a unique and highly specialized cell responsible for creating, activating, and controlling the embryonic genome, as well as supporting basic processes such as cellular homeostasis, metabolism, and cell cycle progression in the early embryo. During oogenesis, the oocyte accumulates a myriad of factors to execute these processes. Oogenesis is critically dependent upon correct oocyte-follicle cell interactions. Disruptions in oogenesis through environmental factors and changes in maternal health and physiology can compromise oocyte quality, leading to arrested development, reduced fertility, and epigenetic defects that affect long-term health of the offspring. Our expanding understanding of the molecular determinants of oocyte quality and how these determinants can be disrupted has revealed exciting new insights into the role of oocyte functions in development and evolution.


Subject(s)
Embryonic Development/physiology , Oocytes/cytology , Oocytes/physiology , Animals , Cell Communication , Cell Polarity , Female , Humans , Male , Mitochondria/physiology , Models, Biological , Mutation , Oogenesis , Ovarian Follicle/cytology , Pregnancy , Pregnancy in Diabetics/physiopathology , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
Biol Reprod ; 79(4): 638-48, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18562704

ABSTRACT

Mouse embryos display a strain-dependent propensity for blastomere cytofragmentation at the two-cell stage. The maternal pronucleus exerts a predominant, transcription-dependent effect on this phenotype, with lesser effects of the ooplasm and the paternal pronucleus. A parental origin effect has been observed as an inequality in the cytofragmentation rate of embryos produced through genetic crosses of reciprocal F(1) hybrid females. To understand the basis for this, we conducted an extensive series of pronuclear transfer studies employing different combinations of inbred and F(1) hybrid maternal and paternal genotypes. We find that the parental origin effect is the result of a transgenerational epigenetic modification, whereby the inherited maternal grandpaternal contribution interacts with the fertilizing paternal genome and the ooplasm. This result indicates that some epigenetic information related to grandparental origins of chromosomes (i.e., imprinting of chromosomes in the mother) is retained through oogenesis and transmitted to progeny, where it affects gene expression from the maternal pronucleus and subsequent embryo phenotype. These results reveal for the first time that mammalian embryonic development can be affected by the epigenotype of at least three individuals. Additionally, we observe a significant suppression of fragmentation by F(1) hybrid ooplasm when it is separated from the F(1) hybrid maternal pronucleus. This latter effect is a striking example of heterosis in the early mammalian embryo, and it provides a new opportunity for examining the molecular mechanisms of heterosis. These results are relevant to our understanding of the mechanisms of epigenetic effects on development and the possible fertility effects of genetic and epigenetic interactions in reproductive medicine.


Subject(s)
Blastocyst/physiology , Genomic Imprinting/physiology , Hybrid Cells/physiology , Animals , Blastocyst/metabolism , Chimera/physiology , Cytokinesis/genetics , Cytokinesis/physiology , Epigenesis, Genetic/physiology , Female , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Models, Biological , Models, Genetic , Phenotype
16.
Biol Reprod ; 78(2): 254-66, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17914069

ABSTRACT

Correct cell cycle regulation is especially challenging at the start of life. Ovulated oocytes must maintain meiotic arrest until fertilization, and then complete meiosis and initiate a series of modified cell divisions without growth. Moreover, myriad key developmental events, such as chromatin remodeling and transcriptional activation of the genome, are coordinated with each other via the cell cycle, particularly passage through the DNA synthesis phase (S Phase). We examined here the expression of more than 30 mRNAs related to cell cycle regulation in rhesus monkey oocytes and embryos and compared the expression of these mRNAs between oocytes and embryos of different developmental potentials. We find that the maternally inherited stores of cell cycle regulatory mRNAs are especially susceptible to disruption in cases of diminished oocyte and embryo quality in the rhesus monkey. In comparison to published mouse array data, we also observed striking species differences in the temporal expression patterns of many of these genes, suggesting that mechanisms of cell cycle control may differ and that the responses of oocytes and embryos to external insults may likewise differ.


Subject(s)
Cell Cycle/genetics , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Genes, cdc , Oocytes/growth & development , Animals , Apoptosis/genetics , Centromere/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclins/genetics , DNA Replication/genetics , Female , Gene Expression , Gene Expression Profiling , Macaca mulatta , Oocytes/metabolism , RNA, Messenger/metabolism , Transcription Factors/genetics
17.
Physiol Genomics ; 31(1): 1-14, 2007 Sep 19.
Article in English | MEDLINE | ID: mdl-17550997

ABSTRACT

Protein degradation via the ubiquitin-proteasome pathway (UPP) plays a key role in diverse aspects of cell physiology and development. In the early embryo, the UPP may play an important role in the transition from maternal to embryonic control of development. Disruptions in the UPP could thus compromise embryo developmental potential. Additionally, species-specific requirements may dictate diverse patterns of regulation of the UPP components. To investigate the expression of UPP components in a nonhuman primate embryo model, to compare expression between a primate and nonprimate species, and to determine whether disruption of this pathway may contribute to reduced developmental potential, we examined the expression of >50 mRNAs encoding UPP components in rhesus monkey oocytes and embryos. We compared this expression between the rhesus monkey and mouse embryo and between rhesus monkey oocytes and embryos of high, intermediate, and low developmental potential. We report here the temporal patterns of UPP gene expression in oocytes and during preimplantation development, including striking differences between the rhesus monkey and mouse. We also report significant differences in UPP gene expression correlating with oocyte and embryo developmental competence and associated with altered regulation of maternally inherited mRNAs encoding these proteins.


Subject(s)
Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Oocytes/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/physiology , Animals , Biological Evolution , Gene Library , Macaca mulatta , Mice , Models, Genetic , Protein Processing, Post-Translational , RNA, Messenger/metabolism , Species Specificity , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...