Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 498, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36627406

ABSTRACT

Considering the amount of global resources and energy consumed, and animal welfare issues associated with traditional meat production, cultured meat production has been proposed as a solution to these problems and is attracting worldwide attention. Cultured meat is produced by culturing/proliferating animal muscle cells in vitro. This process requires significant amounts of culture medium, which accounts to a major portion of the production cost. Furthermore, it is composed of nutrients derived from grains and heterotrophic microorganisms and fetal bovine serum (FBS), which will impact the sustainability of cultured meat in future. Here, we developed a novel medium containing nutrients extracted from microalga and cell-secreted growth factors. First, rat liver epithelial RL34 cells were cultured by adding Chlorella vulgaris extract (CVE) to inorganic salt solution. The supernatant, containing the RL34 cell-secreted growth factors, was used as the conditioned medium (CM). This CM, with CVE added as a nutrient source, was applied to primary bovine myoblast cultures. This serum-free and grain-derived-nutrient-free medium promoted the proliferation of bovine myoblasts, the main cell source for cultured beef. Our findings will allow us to take a major step toward reducing production costs and environmental impacts, leading to an expansion of the cultured meat market.


Subject(s)
Chlorella vulgaris , Microalgae , Animals , Cattle , Cell Culture Techniques , Meat , Culture Media, Conditioned/pharmacology , Cells, Cultured , Mammals
2.
Aging Cell ; 19(10): e13240, 2020 10.
Article in English | MEDLINE | ID: mdl-32959976

ABSTRACT

Pregnancy rate of women decreases with age due to declining quality of oocytes and embryos. However, there is no established method to improve pregnancy rate in aging women. In this study, we identified a senescence-associated secretory phenotype (SASP) factor partially responsible for the decline in embryo implantation potential. Based on microarray analysis using young and aging human embryos at the same morphological grade, 702 genes showed >fivefold increases in aging human blastocysts. Among these genes, C-X-C motif chemokine 5 (CXCL5) showed 7.7-fold increases in aging human blastocysts. However, no-age-dependent changes in expression of the CXCR2, the cognate receptor for CXCL5, were found. In aging mice, Cxcl5 transcript levels were also increased in oocytes and embryos. Treatment of young mouse embryos with CXCL5 decreased implantation rates, together with increased expression of aging markers (P53, P21, Pai-1, and Il-6). Moreover, CXCL5 treatment suppressed trophoblast outgrowth in young mouse blastocysts. Conversely, suppression of CXCL5-CXCR2 signaling in aging mouse embryos using neutralizing antibodies and a receptor antagonist improved the implantation rate, leading to increases in pregnancy and delivery of normal pups. The gene expression pattern of these embryos was comparable to that in young mouse embryos showing enriched cell proliferation-related pathways. In conclusion, we identified CXCL5 as a SASP factor in human and mouse embryos and suppression of CXCL5-CXCR2 signaling during embryo culture improved pregnancy success in aging mice. Future analysis on CXCL5-CXCR2 signaling suppression in human embryos could be the basis to improve embryo development and pregnancy outcome in middle-aged infertile patients.


Subject(s)
Blastocyst/metabolism , Cellular Senescence/physiology , Chemokine CXCL5/metabolism , Receptors, Interleukin-8B/metabolism , Adult , Animals , Female , Humans , Male , Mice , Mice, Inbred ICR , Middle Aged , Signal Transduction
3.
Biol Reprod ; 100(4): 1082-1089, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30561512

ABSTRACT

Heterosis is the beneficial effect of genetical heterogeneity in animals and plants. Although heterosis induces changes in the cells and individual abilities, few reports have described the effect of heterosis on the female reproductive ability during aging. In this study, we investigated the reproductive capability of genetically heterogeneous (HET) mice established by the four-way crossing of C57BL/6N, BALB/c, C3H/He, and DBA/2. We found the HET females naturally and repeatedly produced offspring, even in old age (14-18 months of age). We also found that HET females showed a significantly enlarged body and organ sizes in both youth and old age. In histological analyses, the numbers of primordial follicles, primary follicles, secondary follicles, and corpora lutea were significantly increased in the old ovaries of HET females compared with those in inbred C57BL/6 mice of the same age. In vitro fertilization experiments revealed that aged HET oocytes showed identical rates of fertilization, early development, and birth compared to those of young and old C57BL/6 oocytes. We further found the significantly increased expression of sirtuin genes concomitant with the up-regulation of R-spondin2 in old HET ovaries. These results confirm the novel phenotype, characterized by fertility extension and follicular retention due to heterosis, in old HET females. The HET female will be a valuable model for clarifying the mechanism underlying the effect of heterosis in the field of reproduction.


Subject(s)
Aging , Fertility/genetics , Hybrid Vigor/physiology , Maternal Age , Reproduction/genetics , Aging/physiology , Animals , Crosses, Genetic , Female , Heterozygote , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic
4.
Syst Biol Reprod Med ; 64(1): 3-11, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29224376

ABSTRACT

The ovary is an interesting organ that shows major structural changes within a short period of time during each reproductive cycle. Follicle development is controlled by local paracrine and systemic endocrine factors. Many hormonal and molecular analyses have been conducted to find the mechanisms underlying structural changes in ovaries, However, exact mechanisms still remain to be determined. Recent development of mechanobiology facilitates the understanding on the contribution of physical forces and changes in the mechanical properties of cells and tissues to physiology and pathophysiology. The Hippo signaling pathway is one of the key players in mechanotransduction, providing an understanding of the molecular mechanisms by which cells sense and respond to mechanical signals to regulate cell proliferation and apoptosis for maintaining optimal organ sizes. Our group recently demonstrated the involvement of the Hippo signaling pathway in the regulation of ovarian follicle development. Fragmentation of ovarian cortex into small cubes changed cytoskeletal actin dynamics and induced disruption of the Hippo signaling pathway, leading to the production of CCN growth factors and anti-apoptotic BIRC. These factors, in turn, stimulated secondary follicle growth in vitro and in vivo. In this review, we summarized hormonal regulation of follicular structural changes and further focused on the role of Hippo signaling in the regulation of follicle development. We also suggest a new strategy of infertility treatments in patients with polycystic ovary syndrome and primary ovarian insufficiency based on mechanobiology.


Subject(s)
Gonadal Hormones/metabolism , Mechanotransduction, Cellular , Ovarian Follicle/enzymology , Ovulation , Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Cell Proliferation , Female , Fertility , Hippo Signaling Pathway , Humans , Infertility, Female/enzymology , Infertility, Female/physiopathology , Infertility, Female/therapy , Ovarian Follicle/growth & development , Polycystic Ovary Syndrome/enzymology , Polycystic Ovary Syndrome/physiopathology , Polycystic Ovary Syndrome/therapy , Primary Ovarian Insufficiency/enzymology , Primary Ovarian Insufficiency/physiopathology , Primary Ovarian Insufficiency/therapy , Reproductive Techniques, Assisted
5.
Aging Cell ; 16(6): 1288-1299, 2017 12.
Article in English | MEDLINE | ID: mdl-28857490

ABSTRACT

Irregular menstrual cycles, reduced responses to exogenous hormonal treatments, and altered endocrine profiles (high FSH/high LH/low AMH) are observed in women with increasing age before menopause. In this study, because the granulosa cell-specific Nrg1 knockout mice (gcNrg1KO) presented ovarian and endocrine phenotypes similar to older women, we sought to understand the mechanisms of ovarian aging and to develop a new strategy for improving fertility in older women prior to menopause. In the ovary of 6-month-old gcNrg1KO mice, follicular development was blocked in bilayer secondary follicles and heterogeneous cells accumulated in ovarian stroma. The heterogeneous cells in ovarian stroma were distinguished as two different types: (i) the LH receptor-positive endocrine cells and (ii) actin-rich fibrotic cells expressing collagen. Both the endocrine and fibrotic cells disappeared following long-term treatment with a GnRH antagonist, indicating that the high levels of serum LH induced the survival of both cell types and the abnormal endocrine profile to reduce fertility. Moreover, follicular development to the antral stages was observed with reduced LH and the disappearance of the abnormal stromal cells. Mice treated with the GnRH antagonist regained normal, recurrent estrous cycles and continuously delivered pups for at least for 3 months. We conclude that endocrine and matrix alternations occur within the ovarian stroma with increasing age and that abolishing these alternations resets the cyclical release of LH. Thus, GnRH antagonist treatments might provide a new, noninvasive strategy for improving fertility in a subset of aging women before menopause.


Subject(s)
Aging/genetics , Aromatase/genetics , Fertility/genetics , Neuregulin-1/genetics , Ovary/physiology , Reproduction/genetics , Animals , Aromatase/metabolism , Estrous Cycle/genetics , Female , Horses , Male , Mice , Mice, Knockout
6.
Reproduction ; 153(6): R205-R213, 2017 06.
Article in English | MEDLINE | ID: mdl-28289071

ABSTRACT

The mammalian ovary is an organ that controls female germ cell development, storing them and releasing mature oocytes for transporting to the oviduct. During the fetal stage, female germ cells change from a proliferative state to meiosis before forming follicles with the potential for the growth of surrounding somatic cells. Understanding of molecular and physiological bases of germ cell development in the fetal ovary contributed not only to the elucidation of genetic disorders in primary ovarian insufficiency (POI), but also to the advancement of novel treatments for patients with POI. Accumulating evidence indicates that mutations in NOBOX, DAZL and FIGLAgenes are associated with POI. In addition, cell biology studies revealed the important roles of these genes as essential translational factors for germ cell development. Recent insights into the role of the PI3K (phosphatidylinositol 3-kinase)-Akt signaling pathway in primordial follicle activation allowed the development of a new infertility treatment, IVA (in vitro activation), leading to successful pregnancy/delivery in POI patients. Furthermore, elucidation of genetic dynamics underlying female germ cell development could allow regeneration of oocytes from ES (embryonic stem)/iPS (induced pluripotent stem) cells in mammals. The purpose of this review is to summarize basic findings related to female germ cell development and potential clinical implications, especially focusing on POI etiologies. We also summarize evolving new POI therapies based on IVA as well as oocyte regeneration.


Subject(s)
Cell Differentiation , Germ Cells/pathology , Induced Pluripotent Stem Cells/pathology , Primary Ovarian Insufficiency/etiology , Primary Ovarian Insufficiency/physiopathology , Animals , Female , Humans , Signal Transduction
7.
Endocrinology ; 157(12): 4899-4913, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27732090

ABSTRACT

Adult Leydig cells are derived from proliferating stem/progenitor Leydig cells in the infant testis and subsequent differentiation to steroidogenic cells in adult mice. Leydig cell proliferation in the infant testis occurs primarily in response to increased levels of LH that induce Leydig cell expression of neuregulin 1 (NRG1). Depletion of NRG1 in Nrg1 mutant mice (Nrg1flox;flox;Cyp19a1Cre mice) dramatically reduces Leydig cell proliferation in the infant testes, leading to a reduction of testis weight, epididymial weight, and serum T in the adult mutant mice. The mutant mice are subfertile due to impaired sexual behavior and abnormal elongation of the spermatogenic cells. These defects were reversed by T treatment of the mutant mice in vivo. Furthermore, NRG1 alone induces the proliferation of Leydig cells in cultures of infant (d 10) testes obtained from mutant mice. Collectively these results show that LH induction of NRG1 directly drives the proliferation of Leydig cells in the infant testis, leading to an obligatory number of adult Leydig cells required for the production of sufficient androgen to support and maintain spermatogenesis and sexual behavior of adult male mice.


Subject(s)
Cell Proliferation/physiology , Leydig Cells/metabolism , Neuregulin-1/metabolism , Sexual Behavior, Animal/physiology , Spermatogenesis/physiology , Animals , Cell Proliferation/drug effects , Leydig Cells/cytology , Leydig Cells/drug effects , Male , Mice , Mice, Knockout , Neuregulin-1/genetics , Sexual Behavior, Animal/drug effects , Spermatogenesis/drug effects , Testosterone/pharmacology
8.
Mol Endocrinol ; 28(5): 706-21, 2014 May.
Article in English | MEDLINE | ID: mdl-24650175

ABSTRACT

Neuregulin 1 (NRG1) is induced in granulosa cells by LH and acts on granulosa and cumulus cells during ovulation. In this study, we sought to determine the role of NRG1 in oocyte maturation by generating a granulosa cell-specific Nrg1 knockout mouse (Nrg1(flox/flox);Cyp19a1Cre mice [gcNrg1KO]). In the gcNrg1KO mice, meiosis was induced 2 hours earlier than in control mice. More than 60% of the oocytes in the mutant mice spontaneously re-resumed meiosis beyond the MII stage. The percentage of successful fertilization was comparable in oocytes of both genotypes collected at 14 or 16 hours after human chorionic gonadotropin injection but was significantly lower in oocytes of the gcNrg1KO mice at 18 or 20 hours. The number of pups per litter was significantly decreased in gcNrg1KO mice. To determine the molecular events associated with the abnormal progression of meiosis in the gcNrg1KO mouse oocytes, the defects of cumulus/granulosa cell functions were analyzed. The expression of genes involved in luteinization and cumulus expansion was significantly higher at 2 hours after human chorionic gonadotropin injection in the gcNrg1KO mice; this was related to abnormal activation of protein kinase C (PKC) and phosphorylation of connexin-43 in cumulus cells. Changes in connexin-43 by PKC might lead to early meiotic resumption of oocytes in gcNrg1KO mice. We conclude that NRG1 is induced by LH in mural granulosa cells and exerts an important regulatory role in oocyte meiotic maturation and competence by reducing PKC activation in cumulus cells and preventing premature progression to the MII stage that leads to abnormal fertilization and fertility.


Subject(s)
Cumulus Cells/metabolism , Neuregulin-1/genetics , Oocytes/physiology , Animals , Cells, Cultured , Female , Fertilization , Gap Junctions/metabolism , Humans , Luteinization , Male , Meiosis , Mice, Inbred C57BL , Mice, Knockout , Neuregulin-1/metabolism , Ovarian Follicle/cytology , Promoter Regions, Genetic , Protein Kinase C/metabolism , Transcriptional Activation
9.
Am J Reprod Immunol ; 69(2): 168-79, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23034013

ABSTRACT

PROBLEM: The aim of this study was to find immune-related genes expressed in cumulus cells of ovulated cumulus oocyte complexes (COCs) and to clear the functional roles during fertilization process. METHOD OF STUDY: Ovulated COCs were collected from oviduct 16 hr after the hCG injections followed by eCG priming. The cumulus cells were used for RT-PCR or western blotting study. COCs were also used for in vitro fertilization study. RESULTS: Cramp, Trf, Lyz2, S100a8, and S100a9 were expressed in cumulus cells during ovulation process. The protein levels of CRAMP or transferrin were detected in ovulated COCs and then secreted into hyaluronan-rich matrix. The high dose of these factors reduced the proliferative activity of E. coli; however, the lower levels of them significantly increased the rate of fertilization in in vitro via the induction of sperm capacitation. CONCLUSION: Cumulus-secreted anti-bacterial factors act on sperm to induce sperm capacitation.


Subject(s)
Cumulus Cells/metabolism , Fertilization in Vitro , Oocytes/metabolism , Sperm Capacitation/physiology , Animals , Antimicrobial Cationic Peptides , Calgranulin A/genetics , Calgranulin A/metabolism , Calgranulin B/genetics , Calgranulin B/metabolism , Cathelicidins/genetics , Cathelicidins/metabolism , Chorionic Gonadotropin/pharmacology , Cumulus Cells/cytology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Oocytes/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spermatozoa/physiology , Transferrin/genetics , Transferrin/metabolism
10.
Endocrinology ; 153(8): 3949-59, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22673225

ABSTRACT

Cumulus cell-oocyte complex (COC) expansion is obligatory for LH-induced ovulation and is initiated by LH induction of the epidermal growth factor (EGF)-like factors that mediate the synthesis of the hyaluronan-rich matrix and hyaluronan-stabilizing factors. COC expansion also involves the movement of cumulus cells within the matrix by mechanisms that have not been characterized. We document herein that two proteases, calpain 2 and to a lesser extent calpain 1, are expressed in cumulus cells and that the proteolytic activity of these enzymes is rapidly and significantly increased in COC isolated from human chorionic gonadotropin-induced ovulatory follicles in vivo. Stimulation of calpain activity was associated with proteolytic degradation of paxillin and talin (two components of focal adhesion complexes), cell detachment, and the formation of cell surface bleb-like protrusions. Injection of a calpain inhibitor in vivo reduced 1) human chorionic gonadotropin-stimulated calpain enzyme activity, 2) cell detachment, 3) membrane protrusion formation, and 4) COC expansion by mechanisms that did not alter Has2 expression. During EGF-like factor induction of COC expansion in culture, calpain activity was increased by ERK1/2 and intracellular Ca(2+) signaling pathways. Inhibition of calpain activity in cultured COC blocked cumulus cell detachment, protrusion formation, and the vigorous movement of cumulus cells. As a consequence, COC expansion was impaired. Collectively, these results show that two highly coordinated processes control COC expansion. One process involves the synthesis of the hyaluronan matrix, and the other mediates cumulus cell detachment and movement. The latter are controlled by calpain activation downstream of the EGF receptor activation of the Ca(2+) pathway and ERK1/2 pathways.


Subject(s)
Calpain/metabolism , Cumulus Cells/metabolism , Epidermal Growth Factor/pharmacology , Oocytes/metabolism , Animals , Blotting, Western , Calpain/antagonists & inhibitors , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Female , Fluorescent Antibody Technique , Humans , Mice , Mice, Inbred C57BL , Paxillin/metabolism , Polymerase Chain Reaction
11.
Reprod Toxicol ; 33(3): 322-30, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22285649

ABSTRACT

The biosynthesis of androgens requires multiple steps and during the conversion of pregnenolone to 17α-hydroxypregnenolone and dehydroepiandrosterone (DHEA) by CYP17a1. Acetaldehyde is potentially formed as a by-product in theca cells during antral follicular development. In this study, acetaldehyde level was significantly increased after eCG stimulation and reached a maximum level at 36-h post-eCG. By 48 h, the level of acetaldehyde decreased in association with the induction of aldehyde dehydrogenase (ALDH) type 1 family members. When immature mice were co-injected with the ALDH inhibitor, cyanamide, and eCG, the expression of genes involved in the differentiations of granulosa cells was suppressed and the number of ovulated oocytes was reduced. The in vitro studies showed that ALDH inhibitors prevented FSH-induced granulosa cell differentiation. These results indicate that acetaldehyde is generated as a by-product during steroidogenesis and can exert toxic effects to impair the differentiation of granulosa cells, reduce ovulation and decrease oocyte quality.


Subject(s)
Acetaldehyde/metabolism , Aldehyde Dehydrogenase/metabolism , Ovarian Follicle/physiology , Aldehyde Dehydrogenase/antagonists & inhibitors , Animals , Chorionic Gonadotropin/pharmacology , Cyanamide/pharmacology , Disulfiram/pharmacology , Enzyme Inhibitors/pharmacology , Female , Mice , Mice, Inbred C57BL , Ovarian Follicle/drug effects
12.
Biol Reprod ; 85(5): 1073-82, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21778143

ABSTRACT

During in vitro maturation of porcine cumulus-oocyte complexes (COCs), follicle-stimulating hormone (FSH) increases both prostaglandin E2 (PGE2) production and the expression levels of EGF-like factors. The ligands act on cumulus cells by the autocrine system due to their specific receptors, EP2, EP4, or EGF receptor. When each pathway is suppressed by inhibitors, complete cumulus expansion and oocyte maturation do not occur. In this study, we examined the relationship between both of these pathways in cumulus cells of porcine COCs. When COCs were cultured with FSH, Fshr mRNA expression was immediately decreased within 5 h, whereas Ptger2, Ptger4, and Ptgs2 expression levels were significantly increased in cumulus cells in the culture containing FSH for 5 or 10 h. The PTGS2 inhibitor NS398 significantly suppressed not only PGE2 secretion at any culture time point but also Areg, Ereg, and Tace/Adam17 expression in cumulus cells at 10 and 20 h but not at 1 or 5 h. During the early culture period, phosphorylation of MAPK3 and MAPK1 (MAPK3/1) was not affected by NS398; however, at 10 and 20 h, phosphorylation was suppressed by the drug. Furthermore, down-regulations of MAPK3/1 phosphorylation and expression of the target genes by NS398 was overcome by the addition of either PGE2 or EGF. FSH-induced cumulus expansion and meiotic progression to the MII stage were also suppressed by NS398, whereas these effects were also overcome by addition of either PGE2 or EGF. These results indicated that PGE2 is involved in the sustainable activation of MAPK3/1 in cumulus cells via the induction of EGF-like factor, which is required for cumulus expansion and meiotic maturation of porcine COCs.


Subject(s)
Cell Communication/physiology , Cumulus Cells/metabolism , Dinoprostone/metabolism , Epidermal Growth Factor/metabolism , Feedback, Physiological/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oocytes/metabolism , Animals , Cells, Cultured , Cumulus Cells/cytology , Cumulus Cells/drug effects , Cyclic AMP/metabolism , Cyclooxygenase 2/metabolism , Female , Follicle Stimulating Hormone/pharmacology , In Vitro Techniques , Models, Animal , Nitrobenzenes/pharmacology , Oocytes/cytology , Oocytes/drug effects , Phosphorylation/drug effects , Receptors, FSH/metabolism , Receptors, Prostaglandin E/metabolism , Signal Transduction/physiology , Sulfonamides/pharmacology , Swine
13.
Mol Endocrinol ; 25(1): 104-16, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21047912

ABSTRACT

Epidermal growth factor (EGF)-like factors [amphiregulin (AREG), betacellulin, and epiregulin] are induced by LH and activate the EGF receptor (ERBB1)/ERK1/2 pathway in granulosa cells and cumulus cells of preovulatory follicles to impact ovulation. However, the expression and roles of other ERBB family members and their ligands have not been explored in detail. Herein, we document that two transcripts of the neuregulin (Nrg1) gene are expressed in granulosa cells, and that the type III Nrg1 is induced during ovulation in an ERK1/2 and C/EBPß-dependent manner. Western blotting shows that intact (75 kDa) and secreted (45 kDa) forms of neuregulin 1 (NRG1) are present in the ovary. NRG1 likely binds to ERBB3/ERBB2 complexes that are expressed in granulosa cells and cumulus cells. In cultured granulosa cells, NRG1 selectively stimulates the phosphorylation of AKT/PKB compared to ERK1/2. However, when granulosa cells were cultured with NRG1 and AREG, the phosphorylation of ERK1/2 was markedly enhanced as compared with that by AREG alone. Cotreatment with NRG1 and AREG also increased progesterone production. When cumulus-oocyte complexes (COCs) were cultured with both NRG1 and AREG, the matured oocytes exhibited significantly higher developmental competence as compared with that of oocytes cultured with AREG alone. Collectively, these results document that the expression of type III NRG1 is induced in granulosa cells during ovulation and that NRG1 enhances AREG-induced ERK1/2 phosphorylation in both granulosa cells and cumulus cells. The NRG1 pathway has two roles: one is to enhance AREG-induced progesterone production in granulosa cells, and the other is to regulate oocyte maturation by a cumulus cell-dependent mechanism.


Subject(s)
Cell Differentiation/drug effects , Granulosa Cells/cytology , Luteinizing Hormone/pharmacology , Neuregulin-1/genetics , Oocytes/cytology , Amphiregulin , Animals , Base Sequence , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Cumulus Cells/cytology , Cumulus Cells/drug effects , Cumulus Cells/enzymology , EGF Family of Proteins , Embryonic Development/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation/drug effects , Glycoproteins/pharmacology , Granulosa Cells/drug effects , Granulosa Cells/enzymology , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Ligands , Mice , Molecular Sequence Data , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Oocytes/drug effects , Ovulation/drug effects , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Time Factors
14.
J Reprod Dev ; 56(3): 315-23, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20168049

ABSTRACT

During in vitro maturation of porcine cumulus-oocyte complexes (COCs), progesterone was secreted from cumulus cells and acted on the cumulus cells themselves, which required for cumulus expansion and oocyte maturation. EGF-like factor (amphiregulin, AREG; epiregulin, EREG) and its protease, TACE/ADAM17, are also expressed in cumulus cells, and thereby, soluble EGF domain was acted on the EGF receptor expressed on cumulus cells. In this study, we examined the relationship between progesterone function and EGF-like factor stimuli in cumulus cells of porcine COCs. When COCs were cultured with FSH and LH, Areg, Ereg and Tace/Adam17 were expressed in cumulus cells. Treatment with a progesterone receptor (PGR) antagonist, RU486, did not affect the Areg and Ereg mRNA expression levels at any culture time points. However, the Tace/Adam17 mRNA level, protein level and its activity were significantly suppressed by RU486 at the 30 or 40 h time point. At 20 h of culture, phosphorylation of ERK1/2 and the expressions of target genes (Has2, Tnfaip6 and Ptgs2) were not suppressed by RU486; however, at 40 h, ERK1/2 phosphorylation and the target gene expression levels were significantly downregulated by RU486 in cumulus cells. Furthermore, the negative effects of RU486 at 40 h were overcome by the addition of EGF. These results indicated that the level of TACE/ADAM17 in cumulus cells was regulated by the progesterone-PGR pathway during in vitro maturation of porcine COCs. Therefore, we concluded that the progesterone-induced TACE/ADAM17 leads to production of soluble EGF domain from cumulus cells, which enhances functional changes of cumulus cells and progresses meiotic maturation of oocytes during in vitro maturation of porcine COCs.


Subject(s)
ADAM Proteins/metabolism , Cumulus Cells/cytology , Epidermal Growth Factor/metabolism , Gene Expression Regulation , Oocytes/cytology , Progesterone/metabolism , ADAM17 Protein , Animals , DNA Primers/genetics , Follicle Stimulating Hormone/metabolism , Luteinizing Hormone/metabolism , Mifepristone/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Swine , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL