Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(1)2021 Dec 21.
Article in English | MEDLINE | ID: mdl-35008478

ABSTRACT

The extracellular matrix (ECM) is an essential structure with biological activities. It has been shown that the ECM influences gene expression via cytoskeletal components and the gene expression is dependent upon cell interactions with molecules and hormones. The development of ovarian follicles is a hormone dependent process. The surge in the luteinizing hormone triggers ovulatory changes in oocyte microenvironment. In this review, we discuss how proteolytic cleavage affects formation of cumulus ECM following hormonal stimulation; in particular, how the specific proteasome inhibitor MG132 affects gonadotropin-induced cytoskeletal structure, the organization of cumulus ECM, steroidogenesis, and nuclear maturation. We found that after the inhibition of proteolytic cleavage, gonadotropin-stimulated oocyte-cumulus complexes (OCCs) were without any signs of cumulus expansion; they remained compact with preserved cytoskeletal F-actin-rich transzonal projections through the oocyte investments. Concomitantly, a significant decrease was detected in progesterone secretion and in the expression of gonadotropin-stimulated cumulus expansion-related transcripts, such as HAS2 and TNFAIP6. In agreement, the covalent binding between hyaluronan and the heavy chains of serum-derived the inter-alpha-trypsin inhibitor, essential for the organization of cumulus ECM, was missing.


Subject(s)
Cellular Microenvironment/physiology , Extracellular Matrix/physiology , Oocytes/physiology , Ovarian Follicle/physiology , Animals , Female , Humans , Proteolysis
2.
Int J Mol Sci ; 21(7)2020 Mar 25.
Article in English | MEDLINE | ID: mdl-32218212

ABSTRACT

In the mammalian ovary, the hyaluronan (HA)-rich cumulus extracellular matrix (ECM) organized during the gonadotropin-induced process of oocyte maturation is essential for ovulation of the oocyte-cumulus complex (OCC) and fertilization. Versican is an HA-binding proteoglycan that regulates cell function and ECM assembly. Versican cleavage and function remain to be determined in ovarian follicle. We investigated versican expression in porcine ovarian follicles by real-time (RT)-PCR and western blotting. The aims of the present work were to determine whether 1) versican was produced and cleaved by porcine OCCs during gonadotropin stimulation; 2) these processes were autonomous or required the participation of mural granulosa cells (MGCs); and 3) versican cleavage was involved in the formation or degradation of expanded cumulus ECM. We demonstrate two cleavage products of G1 domain of versican (V1) accumulated in the HA-rich cumulus ECM. One of them, a G1-DPEAAE N-terminal fragment (VG1) of ~70 kDa, was generated from V1 during organization of HA in in vivo and in vitro expanded porcine OCCs. Second, the V1-cleaved DPEAAE-positive form of ~65 kDa was the only species detected in MGCs. No versican cleavage products were detected in OCCs cultured without follicular fluid. In summary, porcine OCCs are autonomous in producing and cleaving V1; the cleaved fragment of ~70 kDa VG1 is specific for formation of the expanded cumulus HA-rich ECM.


Subject(s)
Oocytes/metabolism , Versicans/metabolism , Animals , Cell Differentiation , Cells, Cultured , Epitopes/immunology , Female , Oocytes/cytology , Oocytes/immunology , Swine , Versicans/genetics
3.
Int J Mol Sci ; 19(1)2018 Jan 18.
Article in English | MEDLINE | ID: mdl-29346283

ABSTRACT

Fertilization of the mammalian oocyte requires interactions between spermatozoa and expanded cumulus extracellular matrix (ECM) that surrounds the oocyte. This review focuses on key molecules that play an important role in the formation of the cumulus ECM, generated by the oocyte-cumulus complex. In particular, the specific inhibitors (AG1478, lapatinib, indomethacin and MG132) and progesterone receptor antagonist (RU486) exerting their effects through the remodeling of the ECM of the cumulus cells surrounding the oocyte have been described. After gonadotropin stimulus, cumulus cells expand and form hyaluronan (HA)-rich cumulus ECM. In pigs, the proper structure of the cumulus ECM depends on the interaction between HA and serum-derived proteins of the inter-alpha-trypsin inhibitor (IαI) protein family. We have demonstrated the synthesis of HA by cumulus cells, and the presence of the IαI, tumor necrosis factor-alpha-induced protein 6 and pentraxin 3 in expanding oocyte-cumulus complexes (OCC). We have evaluated the covalent linkage of heavy chains of IαI proteins to HA, as the principal component of the expanded HA-rich cumulus ECM, in porcine OCC cultured in medium with specific inhibitors: AG1478 and lapatinib (both inhibitors of epidermal growth factor receptor tyrosine kinase activity); MG132 (a specific proteasomal inhibitor), indomethacin (cyclooxygenase inhibitor); and progesterone receptor antagonist (RU486). We have found that both RU486 and indomethacin does not disrupt the formation of the covalent linkage between the heavy chains of IαI to HA in the expanded OCC. In contrast, the inhibitors AG1478 and lapatinib prevent gonadotropin-induced cumulus expansion. Finally, the formation of oocyte-cumulus ECM relying on the covalent transfer of heavy chains of IαI molecules to HA has been inhibited in the presence of MG132.


Subject(s)
Cumulus Cells/metabolism , Extracellular Matrix/metabolism , Hyaluronic Acid/metabolism , Oocytes/metabolism , Animals , C-Reactive Protein/metabolism , Cell Adhesion Molecules/metabolism , Cumulus Cells/cytology , Cumulus Cells/drug effects , Extracellular Matrix/drug effects , Female , Mifepristone/pharmacology , Oocytes/cytology , Reproduction/drug effects , Serum Amyloid P-Component/metabolism
4.
Fertil Steril ; 99(6): 1739-48, 2013 May.
Article in English | MEDLINE | ID: mdl-23375200

ABSTRACT

OBJECTIVE: To determine whether inhibition of epidermal growth factor (EGF) receptor tyrosine kinase with lapatinib affects oocyte maturation, expression of the cumulus expansion-associated genes such as tumor necrosis factor alpha-induced protein 6 (TNFAIP6) and prostaglandin-endoperoxide synthase 2 (PTGS2), and synthesis of hyaluronan (HA) and progesterone (P) by porcine oocyte cumulus complexes (OCC). DESIGN: Our work focuses on lapatinib, an orally active small molecule that selectively inhibits the tyrosine kinase domain of both EGF receptor and human EGF receptor 2, and downstream signaling. SETTING: A reproductive biology laboratory. PATIENT(S): Not applicable. INTERVENTION(S): Porcine OCC were cultured in vitro in a medium with FSH/LH in the presence/absence of lapatinib. MAIN OUTCOME MEASURE(S): Methods performed: real-time reverse transcriptase-polymerase chain reaction (PCR), immunofluorescence, RIA. RESULT(S): In FSH/LH-stimulated and expanded cumulus oophorus extracellular matrix, HA was detected with biotinylated HA-binding proteins. However, weaker HA- and weaker cytoplasmic TNFAIP6 were detected were detected in lapatinib-pretreated OCC. The expression of the two cumulus expansion-associated gene transcripts was significantly decreased and synthesis of HA by cumulus cells was reduced. Lapatinib (10 µM) inhibited FSH/LH-induced oocyte meiotic maturation. Progesterone production increased after OCC stimulation with FSH/LH and was significantly decreased by lapatinib (10 µM). CONCLUSION(S): Lapatinib inhibits oocyte maturation and reduces expression of cumulus expansion-associated transcripts, and synthesis of HA and P in OCC cultured in vitro in FSH/LH-supplemented medium.


Subject(s)
Cell Differentiation/drug effects , Cumulus Cells/drug effects , Follicle Stimulating Hormone/pharmacology , Growth Inhibitors/pharmacology , Meiosis/drug effects , Oocytes/drug effects , Quinazolines/pharmacology , Animals , Cell Differentiation/physiology , Cells, Cultured , Cumulus Cells/cytology , Female , Lapatinib , Meiosis/physiology , Oocytes/cytology , Swine
5.
Mol Reprod Dev ; 78(6): 391-402, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21520325

ABSTRACT

Several lines of evidence suggest that in mice the activation of SMAD2/3 signaling by oocyte secreted factors, together with epidermal growth factor receptor (EGFR) activation, is essential to induce cumulus expansion. Here we show that inhibition of EGFR kinase in follicle stimulating hormone (FSH)-stimulated porcine oocyte-cumulus cell complex (OCCs) strongly decreases hyaluronan (HA) synthesis and its retention in the matrix, as well as progesterone synthesis. Although porcine cumulus cells undergo expansion independently of oocytes, we use biochemical and gene expression analyses to show that they do require activation of SMAD2/3 for optimal stimulation of HA synthesis and proteins involved in the organization of this polymer in the expanded matrix. Furthermore, FSH-induced progesterone synthesis by porcine cumulus cells was increased by blocking SMAD2/3 activation. In conclusion, these results support the hypothesis that an FSH-EGF autocrine loop is active in porcine OCCs, and provide the first evidence that the SMAD2/3 signaling pathway is induced by paracrine/autocrine factors in porcine cumulus cells and is involved in the control of both cumulus expansion and steroidogenesis.


Subject(s)
Cumulus Cells/metabolism , ErbB Receptors/metabolism , Hyaluronic Acid/biosynthesis , Isoquinolines/pharmacology , Oocytes/enzymology , Progesterone/biosynthesis , Pyridines/pharmacology , Pyrroles/pharmacology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Animals , Benzamides/pharmacology , C-Reactive Protein/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/metabolism , Dioxoles/pharmacology , Epidermal Growth Factor/metabolism , ErbB Receptors/antagonists & inhibitors , Female , Follicle Stimulating Hormone/metabolism , Gene Expression Regulation, Developmental/drug effects , Glucuronosyltransferase/antagonists & inhibitors , Glucuronosyltransferase/metabolism , Meiosis/drug effects , Mice , Oocytes/physiology , Quinazolines/pharmacology , Serum Amyloid P-Component/metabolism , Signal Transduction/drug effects , Smad2 Protein/antagonists & inhibitors , Smad3 Protein/antagonists & inhibitors , Swine , Tyrphostins/pharmacology
6.
Reproduction ; 141(4): 425-35, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21239527

ABSTRACT

The aim of this work was to assess the FSH-stimulated expression of epidermal growth factor (EGF)-like peptides in cultured cumulus-oocyte complexes (COCs) and to find out the effect of the peptides on cumulus expansion, oocyte maturation, and acquisition of developmental competence in vitro. FSH promptly stimulated expression of amphiregulin (AREG) and epiregulin (EREG), but not betacellulin (BTC) in the cultured COCs. Expression of AREG and EREG reached maximum at 2 or 4 h after FSH addition respectively. FSH also significantly stimulated expression of expansion-related genes (PTGS2, TNFAIP6, and HAS2) in the COCs at 4 and 8 h of culture, with a significant decrease at 20 h of culture. Both AREG and EREG also increased expression of the expansion-related genes; however, the relative abundance of mRNA for each gene was much lower than in the FSH-stimulated COCs. In contrast to FSH, AREG and EREG neither stimulated expression of CYP11A1 in the COCs nor an increase in progesterone production by cumulus cells. AREG and EREG stimulated maturation of oocytes and expansion of cumulus cells, although the percentage of oocytes that had reached metaphase II was significantly lower when compared to FSH-induced maturation. Nevertheless, significantly more oocytes stimulated with AREG and/or EREG developed to blastocyst stage after parthenogenetic activation when compared to oocytes stimulated with FSH alone or combinations of FSH/LH or pregnant mares serum gonadotrophin/human chorionic gonadotrophin. We conclude that EGF-like peptides do not mimic all effects of FSH on the cultured COCs; nevertheless, they yield oocytes with superior developmental competence.


Subject(s)
Cell Proliferation/drug effects , Cumulus Cells/drug effects , Gonadotropins/pharmacology , Oocytes/drug effects , Peptide Fragments/pharmacology , Swine , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Cumulus Cells/metabolism , Cumulus Cells/physiology , Embryo Culture Techniques , Embryonic Development/drug effects , Embryonic Development/genetics , Epidermal Growth Factor/chemistry , Epidermal Growth Factor/pharmacology , Female , Follicle Stimulating Hormone/pharmacology , Gene Expression Profiling , Oocytes/metabolism , Oocytes/physiology , Oogenesis/drug effects , Oogenesis/genetics , Parthenogenesis/drug effects , Parthenogenesis/genetics , Parthenogenesis/physiology , Peptide Fragments/chemistry , Swine/genetics , Swine/metabolism , Swine/physiology
7.
Mol Reprod Dev ; 76(8): 751-61, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19382213

ABSTRACT

We show in the present study that freshly isolated pig cumulus-oocyte complexes (COCs) display a limited response to LH, as assessed by the expression of hyaluronan synthase 2 (Has2) mRNA, activation of protein kinase A (PKA), production of hyaluronic acid (HA) and progesterone, cumulus cell expansion and resumption of meiosis. These data indicate that freshly isolated COCs do not possess a sufficient number of functional LH receptors (LHR). However, the expression of Lhr significantly increased during the culture of COCs in vitro in a medium supplemented with FSH. Assuming that the effect of FSH on LHR induction is mediated via cAMP signaling pathways, we developed a new culture system, in which the COCs were pre-cultured for 72 hr in a medium supplemented with dbcAMP. The pre-cultured COCs remained in the germinal vesicle stage, their cumulus investment underwent a dramatic increase in size and gap junctions between the cumulus cells were preserved. The stimulation of such COCs with either FSH or LH led to the resumption and completion of meiosis, activation of PKA, expression of Has2, synthesis of large amounts of HA and progesterone, and extensive expansion of cumulus cells. We conclude that the formation of functional LHR is stimulated in cumulus cells during the culture in vitro in a cAMP-dependent pathway. The dbcAMP-treated COCs thus represent a new model in which the resumption of meiosis and cumulus expansion can be induced exclusively by the action of recombinant LH.


Subject(s)
Cumulus Cells/metabolism , Oocytes/growth & development , Ovarian Follicle/metabolism , Receptors, LH/biosynthesis , Analysis of Variance , Animals , Bucladesine/pharmacology , Cell Culture Techniques/methods , Cumulus Cells/cytology , Cumulus Cells/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Follicle Stimulating Hormone/pharmacology , Gene Expression/drug effects , Glucuronosyltransferase/biosynthesis , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Hyaluronic Acid/metabolism , Luteinizing Hormone/pharmacology , Oocytes/cytology , Oocytes/drug effects , Ovarian Follicle/drug effects , Progesterone/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, LH/genetics , Receptors, LH/metabolism , Swine
8.
J Reprod Dev ; 55(3): 231-5, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19194062

ABSTRACT

It has been shown previously that tumor necrosis factor alpha-induced protein 6 (TNFAIP6) is essential for formation of the cumulus extracellular matrix and female fertility. Therefore, we studied the expression of TNFAIP6 mRNA in porcine preovulatory follicles. In addition, we asked whether the expression of TNFAIP6 mRNA changes in mural granulosa cells (MGCs) during the periovulatory period or after culture of oocyte-cumulus complexes (OCCs) or MGCs in vitro. Mural granulosa cells obtained from follicles on day 12 (D 12) and day 15 (D 15) of the estrous cycle, eCG-stimulated follicles, follicles at 4-32 h after hCG stimulation and MGCs and OCCs obtained from immature gilts and cultured for 0-44 h in vitro with gonadotropins were used for extraction of total RNA and assessment of the relative abundance (RA) of TNFAIP6 mRNA by reverse transcription-polymerase chain reaction (RT-PCR). The levels of TNFAIP6 mRNA were low in the follicles on D 12 and D 15 of the estrous cycle and at 66 h after eCG stimulation but were significantly increased at 4 h after hCG. The high level of TNFAIP6 expression was maintained until 16 h after hCG stimulation and gradually decreased at 24 and 32 h after hCG. During in vitro culture, FSH/LH-induced TNFAIP6 mRNA was expressed in both OCCs and MGCs in a similar temporal pattern as seen in vivo. We conclude that TNFAIP6 expression in the pig, like other species, increases in preovulatory follicles following the LH (hCG) surge. The OCC and MGC display similar patterns of TNFAIP6 expression under both in vivo and in vitro conditions.


Subject(s)
Cell Adhesion Molecules/genetics , Ovarian Follicle/metabolism , Ovulation/genetics , Swine/genetics , Animals , Cell Adhesion Molecules/metabolism , Cells, Cultured , Female , Gene Expression , Ovulation/metabolism , RNA, Messenger/metabolism , Swine/metabolism , Tissue Distribution , Tumor Necrosis Factor-alpha/pharmacology
9.
Toxicol In Vitro ; 23(3): 371-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19162163

ABSTRACT

In most mammals, before ovulation, cumulus cells synthesize a large amount of hyaluronan (HA) that is organized into an extracellular matrix (ECM), which provides an essential microenvironment for in vivo oocyte fertilization. This process is called cumulus expansion. The present study assessed effects of selected endocrine disruptors (bisphenol A, BPA; 4-chloro-3-methyl phenol, CMP; di(2-ethylhexyl) phthalate, DEHP; and benzyl butyl phthalate, BBP) in a range of 100pM-100microM, on follicle-stimulating hormone (FSH)-induced meiotic maturation and cumulus expansion of porcine oocyte-cumulus complexes (OCC) cultured in vitro. Moreover, FSH-stimulated production of hyaluronic acid (HA) and progesterone by cumulus cells was measured. Both phenols, BPA and CMP (100microM), significantly affected meiotic maturation of oocytes. The number of oocytes that underwent germinal vesicle breakdown (GVBD) (78.7% and 72.4%, respectively) as well as the rate of oocytes that reached metaphase II stage (MII) (50% and 53.6%, respectively) after 44h culture were decreased compared to control (89.6% for GVBD and 81.5% for MII). FSH-stimulated expansion of cumulus was altered by the highest concentration of BPA and CMP (70% and 64%, respectively vs. 80.3% in control). Although BPA did not alter FSH-stimulated HA synthesis by cumulus cells, its incorporation within the complex was reduced to a half of control value. Progesterone production by OCC was significantly changed in the presence of BPA or DEHP. Finally, our results provide valuable information that oocyte meiotic progression was adversely affected during in vitro culture with endocrine disruptors.


Subject(s)
Cumulus Cells/drug effects , Endocrine Disruptors/toxicity , Hyaluronic Acid/biosynthesis , Meiosis/drug effects , Oocytes/drug effects , Progesterone/biosynthesis , Animals , Benzhydryl Compounds , Cumulus Cells/metabolism , Female , Meiosis/physiology , Mitosis/drug effects , Mitosis/physiology , Oocytes/growth & development , Oocytes/metabolism , Phenols/toxicity , Phthalic Acids/toxicity , Swine
10.
Biol Reprod ; 78(5): 903-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18256330

ABSTRACT

We have previously shown that the heavy chains (HCs) of inter-alpha-trypsin inhibitor (IalphaI) become covalently linked to hyaluronan (HA) during in vivo and in vitro expansion of porcine oocyte-cumulus cell complexes (OCCs). We have now studied by immunoblotting the synthesis of tumor necrosis factor alpha-induced protein 6 (TNFAIP6), which is essential for catalyzing this reaction in expanding mouse OCCs. Expanding OCCs were collected from preovulatory follicles of naturally cycling pigs and also after in vitro culture (24 or 42 h) in medium supplemented with FSH and pig serum. After isolation, OCCs were treated with Streptomyces hyaluronidase or Chondroitinase ABC. Matrix, cell pellet, and total extracts were analyzed by Western blotting. A band of about 35 kDa and a doublet of about 120 kDa, corresponding to the molecular weight of the native and HC-linked forms of TNFAIP6, respectively, were detected by a rabbit anti-human TNFAIP6 polyclonal antibody in matrix extracts of expanded cumuli. Moreover, we found by using a cell-free assay that porcine follicular fluid collected from follicles at 24 h after hCG stimulation contains HC-HA coupling activity. This activity was abolished by the rat anti-human monoclonal antibody A38, which has an epitope within the Link module domain of TNFAIP6. These experiments suggest that free TNFAIP6 protein was present in follicular fluid aspirated from porcine follicles 24 h after hCG stimulation. In contrast to mouse, we show that the A38 monoclonal antibody does not affect in vitro cumulus expansion of porcine OCCs.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cell Adhesion Molecules/metabolism , Follicular Phase/metabolism , Ovarian Follicle/metabolism , Alpha-Globulins/metabolism , Animals , Antibodies, Monoclonal/immunology , Cell Adhesion Molecules/immunology , Cells, Cultured , Epitopes/metabolism , Female , Follicular Fluid/metabolism , Hyaluronic Acid/metabolism , Mice , Mice, Inbred Strains , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Swine , Time Factors
11.
Biol Reprod ; 78(1): 115-26, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17942798

ABSTRACT

The resumption of oocyte meiosis in mammals encompasses the landmark event of oocyte germinal vesicle (GV) breakdown (GVBD), accompanied by the modification of cell-to-cell communication and adhesion between the oocyte and surrounding cumulus cells. The concomitant cumulus expansion relies on microfilament-cytoskeletal remodeling and extracellular matrix (ECM) deposition. We hypothesized that this multifaceted remodeling event requires substrate-specific proteolysis by the ubiquitin-proteasome pathway (UPP). We evaluated meiotic progression, cytoskeletal dynamics, and the production of cumulus ECM in porcine cumulus-oocyte complexes (COCs) cultured with or without 10-200 microM MG132, a specific proteasomal inhibitor, for the first 22 h of in vitro maturation, followed by 22 h of culture with or without MG132. Treatment with 10 microM MG132 arrested 28.4% of oocytes in GV stage (vs. 1.3% in control), 43.1% in prometaphase I, and 16.2% in metaphase I, whereas 83.7% of control ova reached metaphase II (0% of MG132 reached metaphase II). The proportion of GV-stage ova increased progressively to >90% with increased concentration of MG132 (20-200 microM). Furthermore, MG132 blocked the extrusion of the first polar body and degradation of F-actin-rich transzonal projections (TZP) interconnecting cumulus cells with the oocyte. The microfilament disruptor cytochalasin E (CE) prevented cumulus expansion but accelerated the breakdown of TZPs. Ova treated with a combination of 10 microM MG132 and 10 microM CE underwent GVBD, despite the inhibition of proteasomal activity. However, 90.0% of cumulus-free ova treated with 10 microM MG132 remained in GV stage, compared with 16.7% GV ova in control. Cumulus expansion, retention of hyaluronic acid, and the deposition of cumulus ECM relying on the covalent transfer of heavy chains of inter-alpha trypsin inhibitor (IalphaI) were also inhibited by MG132. Cumulus expansion in control COCs was accompanied by the degradation of ubiquitin-C-terminal hydrolase L3, an important regulator of UPP. RAC1, a UPP-controlled regulator of actin polymerization was maintained at steady levels throughout cumulus expansion. We conclude that proteasomal proteolysis has multiple functions in the progression of oocyte meiosis beyond GV and metaphase I stage, polar body extrusion, and cumulus expansion.


Subject(s)
Cumulus Cells/physiology , Meiosis/physiology , Oocytes/physiology , Proteasome Endopeptidase Complex/metabolism , Swine , Actin Cytoskeleton/metabolism , Animals , Cells, Cultured , Cumulus Cells/cytology , Cumulus Cells/drug effects , Cytoplasm/metabolism , Cytoskeleton/drug effects , Dose-Response Relationship, Drug , Extracellular Matrix , Female , Gonadotropins/pharmacology , Leupeptins/pharmacology , Meiosis/drug effects , Oocytes/cytology , Oocytes/drug effects , Ovarian Follicle
12.
Biol Reprod ; 76(6): 1016-24, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17329596

ABSTRACT

The purpose of the present study was to elucidate signaling pathways by which insulin like-growth factor 1 (IGF1) promotes FSH-stimulated synthesis and retention of hyaluronic acid (HA) in pig oocyte-cumulus complexes (OCCs) cultured in serum-free medium. We found that IGF1 had no effects on FSH-stimulated production of cAMP and activation of protein kinase A in the OCCs. Immunoblotting with phospho-specific antibodies showed that FSH moderately phosphorylated v-akt murine thymoma viral oncogene homolog (AKT) and mitogen-activated kinase 3 and 1 (MAPK3/1) in cumulus cells. The exposure of OCCs to both FSH and IGF1 resulted in a significant (P < 0.05) increase in AKT and MAPK3/1 phosphorylation. An inhibitor of phosphoinositide-3-kinase (PIK3), LY 294002, significantly (P < 0.05) reduced the IGF1-enhanced phosphorylation of AKT, and inhibitors of AKT (SH6) and MAPK3/1 (U0126) significantly (P < 0.05) decreased the synthesis and retention of HA stimulated by concomitant exposure of OCCs to both FSH and IGF1. The IGF1-promoted synthesis of HA was not accompanied by an increase in the relative abundance of hyaluronan synthase 2 (HAS2) mRNA in the cumulus cells. We conclude that IGF1 promotes the FSH-stimulated synthesis and retention of HA in pig OCCs by PIK3/AKT- and MAPK3/1-dependent mechanisms.


Subject(s)
Granulosa Cells/drug effects , Hyaluronic Acid/biosynthesis , Hyaluronic Acid/metabolism , Insulin-Like Growth Factor I/pharmacology , Oocytes/drug effects , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP/biosynthesis , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Follicle Stimulating Hormone/pharmacology , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Granulosa Cells/cytology , Granulosa Cells/metabolism , Hyaluronan Synthases , Mitogen-Activated Protein Kinase 3/metabolism , Oocytes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Swine
13.
Biol Reprod ; 71(6): 1838-43, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15286040

ABSTRACT

Previous studies have shown that the heavy chains (HCs) of serum-derived inter-alpha-trypsin inhibitor (IalphaI) molecules become covalently linked to hyaluronan (HA) during in vivo mouse cumulus expansion and significantly contribute to cumulus matrix organization. Experiments with mice suggest that the incorporation of such proteins in cumulus matrix appears to be rather complex, involving LH/hCG-induced changes in blood-follicle barrier and functional cooperation between cumulus cells, granulosa cells, and oocyte within the follicle. We demonstrate here that HC-HA covalent complexes are formed during in vivo porcine cumulus expansion as well. Western blot analysis with IalphaI antibody revealed that follicular fluids from medium-sized follicles and those from large follicles unstimulated with hCG contain high levels of all forms of IalphaI family members present in pig serum. The same amount of HCs were covalently transferred from IalphaI molecules to HA when pig oocyte-cumulus complexes (OCCs) were stimulated in vitro with FSH in the presence of pig serum or follicular fluid from unstimulated or hCG-stimulated follicles. In addition, HC-HA coupling activity was stimulated in cumulus cells by FSH treatment also in the absence of oocyte. Collectively, these results indicate that IalphaI molecules can freely cross the blood follicle barrier and that follicular fluid collected at any stage of folliculogenesis can be successfully used instead of serum for improving OCC maturation. Finally, pig cumulus cells show an autonomous ability to promote the incorporation of IalphaI HCs in the cumulus matrix.


Subject(s)
Alpha-Globulins/chemistry , Alpha-Globulins/metabolism , Hyaluronic Acid/metabolism , Oocytes/metabolism , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Animals , Blood , Chorionic Gonadotropin/administration & dosage , Chorionic Gonadotropin/pharmacology , Female , Follicle Stimulating Hormone/pharmacology , Follicular Fluid/metabolism , Injections , Oocytes/physiology , Ovarian Follicle/physiology , Swine , Tissue Culture Techniques
14.
Biol Reprod ; 71(4): 1290-5, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15189836

ABSTRACT

We have shown previously that porcine cumulus and mural granulosa cells produce a factor that is very similar, if not identical, to the oocyte-derived cumulus expansion-enabling factor (CEEF). Because growth differentiation factor 9 (GDF9) is the most likely candidate for the CEEF, in the present study we tested the hypothesis that GDF9 is expressed not only in oocytes in the pig but also in somatic follicular cells. In addition, we asked whether the relative abundance (RA) of GDF9 mRNA changes in oocytes and/or follicular cells during the periovulatory period or culture of oocyte-cumulus complexes (OCCs) in vitro. Denuded oocytes, OCCs, cumulus, and mural granulosa cells were isolated from growing and preovulatory follicles. Total RNA was extracted from the cells, and reverse transcription-polymerase chain reaction (RT-PCR) was carried out using specific oligonucleotide primers. The RT-PCR resulted in amplification of a product of expected size (277 base pairs) in samples prepared from all follicular cell types. The identity of the RT-PCR products with GDF9 was confirmed by analysis of their nucleotide sequence, which was 88% and 91% identical to human and ovine GDF9, respectively. The RA of GDF9 mRNA in the somatic follicular cells was approximately fourfold lower than in oocytes. Assessment of the RA of GDF9 mRNA during the periovulatory period and during culture and expansion of OCCs in vitro revealed that it remained stable in oocytes and mural granulosa cells and decreased significantly in expanding cumulus cells. We conclude that GDF9 mRNA can be produced by somatic follicular cells in the pig and that cumulus expansion is not preceded or accompanied by an increase in the RA of GDF9 mRNA in any of the tested cell types.


Subject(s)
Estrous Cycle/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Oocytes/metabolism , Ovarian Follicle/metabolism , RNA, Messenger/metabolism , Analysis of Variance , Animals , Bone Morphogenetic Protein 15 , Female , Follicular Phase/physiology , Granulosa Cells/metabolism , Growth Differentiation Factor 9 , Intercellular Signaling Peptides and Proteins/genetics , Ovarian Follicle/cytology , Swine
15.
Biol Reprod ; 68(3): 797-803, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12604628

ABSTRACT

We have recently shown that epidermal growth factor (EGF) strongly stimulates expansion of porcine oocyte-cumulus complexes (OCCs) isolated from large follicles (>6 mm) and does not promote expansion of OCCs from small (3-4-mm) follicles. In order to elucidate the role of EGF in OCCs expansion, in the present study, we first examined the presence of EGF receptors (EGFRs) in cumulus cells isolated from follicles of different sizes. Surprisingly, immunoblotting showed that cumulus cells obtained from all follicular size categories contained similar amounts of EGFR protein. On the other hand, we found a dramatic difference in the pattern of protein tyrosine phosphorylation in a comparison of cumulus cells isolated from small and large follicles treated by EGF. Furthermore, tyrosine-phosphorylated EGFR was specifically immunoprecipitated with antiphosphotyrosine antibodies from EGF-treated cumulus cells isolated from the large follicles. This result strongly indicates that only OCCs from the large follicles contain mature EGFRs that are capable of becoming activated by EGF. Remarkably, preincubation of cumulus cells from small follicles (3-4 mm) with FSH strongly increased EGF-stimulated tyrosine phosphorylation to levels comparable with OCCs from large follicles. The FSH-dependent activation of EGFRs was beneficial for expansion of OCCs isolated from the small follicles since OCCs treated sequentially by FSH (3 h) and EGF (1 h) underwent expansion significantly better then OCCs cultured in FSH or EGF alone. We conclude that a FSH-dependent pathway has an important role in the maturation of the EGFR in cumulus cells and that activation of EGFR-dependent signaling is sufficient to induce expansion.


Subject(s)
ErbB Receptors/metabolism , Oocytes/enzymology , Ovarian Follicle/enzymology , Swine/metabolism , Animals , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/metabolism , ErbB Receptors/antagonists & inhibitors , Female , Follicle Stimulating Hormone/metabolism , Granulosa Cells/enzymology , Hyaluronic Acid/metabolism , Immunoblotting/veterinary , Oocytes/physiology , Ovarian Follicle/physiology , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Tyrphostins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...