Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Clin Epigenetics ; 13(1): 61, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33757590

ABSTRACT

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is the most common genetic disease of the cardiac muscle, frequently caused by mutations in MYBPC3. However, little is known about the upstream pathways and key regulators causing the disease. Therefore, we employed a multi-omics approach to study the pathomechanisms underlying HCM comparing patient hearts harboring MYBPC3 mutations to control hearts. RESULTS: Using H3K27ac ChIP-seq and RNA-seq we obtained 9310 differentially acetylated regions and 2033 differentially expressed genes, respectively, between 13 HCM and 10 control hearts. We obtained 441 differentially expressed proteins between 11 HCM and 8 control hearts using proteomics. By integrating multi-omics datasets, we identified a set of DNA regions and genes that differentiate HCM from control hearts and 53 protein-coding genes as the major contributors. This comprehensive analysis consistently points toward altered extracellular matrix formation, muscle contraction, and metabolism. Therefore, we studied enriched transcription factor (TF) binding motifs and identified 9 motif-encoded TFs, including KLF15, ETV4, AR, CLOCK, ETS2, GATA5, MEIS1, RXRA, and ZFX. Selected candidates were examined in stem cell-derived cardiomyocytes with and without mutated MYBPC3. Furthermore, we observed an abundance of acetylation signals and transcripts derived from cardiomyocytes compared to non-myocyte populations. CONCLUSIONS: By integrating histone acetylome, transcriptome, and proteome profiles, we identified major effector genes and protein networks that drive the pathological changes in HCM with mutated MYBPC3. Our work identifies 38 highly affected protein-coding genes as potential plasma HCM biomarkers and 9 TFs as potential upstream regulators of these pathomechanisms that may serve as possible therapeutic targets.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/physiopathology , Carrier Proteins/genetics , DNA Methylation , Gene Expression , Genes, Homeobox , Histones/genetics , Humans , Mutation , Transcriptome
2.
Bratisl Lek Listy ; 120(1): 46-51, 2019.
Article in English | MEDLINE | ID: mdl-30685992

ABSTRACT

OBJECTIVES: For the first time we used targeted next-generation sequencing to detect candidate pathogenic variants in Slovak cardiomyopathy patients. BACKGROUND: Targeted next-generation sequencing is considered to be the best practice in genetic diagnostics of cardiomyopathies. However, in Slovakia, with high cardiomyopathies prevalence of 1/440, the current diagnostic tests are still based on Sanger sequencing of a few genes. Consequently, little is known about the exact contribution of pathogenic variants in known cardiomyopathy genes in Slovak patients. METHODS: We used a panel of 46 known cardiomyopathy-associated genes to detect genetic variants in 16 Slovak cardiomyopathy patients (6 dilated, 8 hypertrophic, 2 non-compaction subtypes). RESULTS: We identified candidate pathogenic variants in 11 of 16 patients (69 %). Genes with higher count of candidate pathogenic variants were MYBPC3, MYH and TTN, each with 3 different variants. Seven variants ACTC1 (c.329C>T), ANKRD1 (c.683G>T), MYH7 (c.1025C>T), PKP2 (c.2003delA), TTN (c.51655C>T, c.84841G>T, c.101874_101881delAGAATTTG) have been detected for the first time and might represent Slovak-specific genetic cause. CONCLUSIONS: We have performed genetic testing of previously untested Slovak cardiomyopathy patients using next-generation sequencing cardiomyopathy gene panel. Given the high percentage of candidate pathogenic variants it should be recommended to implement this method into routine genetic diagnostic practice in Slovakia (Tab. 4, Ref. 39).


Subject(s)
Cardiomyopathies , Cardiomyopathy, Hypertrophic , High-Throughput Nucleotide Sequencing , Cardiomyopathies/diagnosis , Cardiomyopathies/drug therapy , Cardiomyopathies/genetics , Cardiomyopathy, Hypertrophic/diagnosis , Cardiomyopathy, Hypertrophic/drug therapy , Cardiomyopathy, Hypertrophic/genetics , Genetic Testing , Humans , Slovakia
3.
J Physiol Pharmacol ; 68(5): 683-691, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29375042

ABSTRACT

Bone morphogenetic protein-15 (BMP-15), an oocyte-derived growth factor, has been shown to play integral roles in regulation of ovarian follicular function in mammals. Despite the recognition of the physiological importance of the BMP system in regulation of gonadotropin action in the ovary, molecular mechanisms of BMP-15 effect on oocyte and somatic follicular cell functions remain poorly understood. The objective of this study was to determine the effect of BMP-15 on the FSH/LH-stimulated synthesis of hyaluronan (HA) by oocyte cumulus complexes (OCC) and progesterone by OCC and granulosa cells (GC) in the presence or absence of serum using primary porcine cultures. In addition, the effect of BMP-15 on oocyte maturation- and steroidogenesis-related transcripts after 4, 8, 16, and 24 hours of cultivation was evaluated using real-time RT-PCR. We demonstrated that the FSH/LH-induced cumulus expansion was accompanied by a significant increase in CD44, PTGS2, CYP11A1 (at 4 h) and AREG, HAS2, TNFAIP6, STAR (at 8 h) mRNAs. While FSH/LH-stimulated total HA synthesis by OCC was not affected by BMP-15 in serum-supplemented medium, its retention within the complex was significantly increased after the action of BMP-15 in comparison to FSH/LH alone (P < 0.001; 65% versus 35%, respectively). Moreover, we detected a significant increase in the expression of AREG and TNFAIP6 (both at 16 h), and CYP11A1 (at 24 h) in FSH/LH-stimulated OCC due to the action of BMP-15 compared to complexes cultured only with FSH/LH. In the presence of serum, BMP-15 markedly increased FSH/LH-stimulated progesterone secretion by OCC (about 69%) and induced a significant decrease in FSH/LH-induced progesterone release by GC (about 35%) compared to FSH/LH alone. The present results indicate that the addition of BMP-15 to the gonadotropin-stimulated OCC cultured in serum-supplemented medium might improve oocyte-cumulus maturation.


Subject(s)
Bone Morphogenetic Protein 15/pharmacology , Gonadotropins/pharmacology , Hyaluronic Acid/biosynthesis , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Progesterone/biosynthesis , Animals , Cells, Cultured , Female , Swine
4.
Domest Anim Endocrinol ; 56: 29-35, 2016 07.
Article in English | MEDLINE | ID: mdl-26986845

ABSTRACT

It has been previously shown that multimeric pentraxin 3 (PTX3) is a key component of the cumulus oophorus extracellular matrix (ECM) in mice. In response to the ovulatory LH surge, the cumulus cells assemble a unique ECM that envelopes the oocyte and cumulus cell complex. Importantly, cumuli from PTX3(-/-) mice were defective in their ECM organization and their fertility was impaired. It has been demonstrated that tumor necrosis factor alpha-induced protein 6 catalyzes the formation of heavy chains of (inter-alpha-trypsin inhibitor) -hyaluronan complexes and these are then cross-linked via PTX3. This process is tightly regulated and requires the proteins to meet/interact in the correct order. Finally, in this way, the above-listed proteins form the cumulus oophorus ECM. We investigated whether PTX3 is expressed in the porcine preovulatory follicle. Porcine oocyte-cumulus complexes (OCC) and mural granulosa cells (MGC) from gilts were obtained either after stimulation in vivo with eCG/hCG (4, 8, 16, 24, and 32 h) or culture in vitro (4, 24, and 44 h) in FSH/LH-supplemented medium. The methods performed were real-time reverse transcriptase-polymerase chain reaction, Western blot analysis, and immunostaining. The expression of PTX3 transcripts was significantly increased 24 h after either in vivo hCG stimulation or in vitro FSH/LH treatment in both OCC and MGC. Western blot analysis with PTX3 antibody revealed that not only matrix extracts from in vivo-stimulated gilts contain high levels of PTX3 protein but also matrix extracts of FSH/LH-stimulated OCC cultured in medium supplemented either with follicular fluid or with porcine serum. The localization of PTX3 in the cumulus oocyte complex was confirmed by immunostaining. In conclusion, PTX3 is produced by porcine OCC and MGC both in vivo and in vitro with gonadotropin stimuli inducing cumulus expansion.


Subject(s)
C-Reactive Protein/genetics , Gene Expression/drug effects , Gonadotropins/pharmacology , Granulosa Cells/metabolism , Oocytes/metabolism , Serum Amyloid P-Component/genetics , Sus scrofa/metabolism , Animals , C-Reactive Protein/analysis , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Culture Media , Cumulus Cells/chemistry , Cumulus Cells/metabolism , Female , Follicle Stimulating Hormone/pharmacology , Gonadotropins, Equine/pharmacology , Luteinizing Hormone/pharmacology , Oocytes/chemistry , RNA, Messenger/analysis , Serum Amyloid P-Component/analysis
5.
Endocr Regul ; 49(1): 37-45, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25687679

ABSTRACT

It has been shown that following endogenous gonadotropin surge, oocyte-cumulus complexes (OCC) synthesize hyaluronan (HA) in a process called cumulus expansion. During this process, HA associates with proteins and proteoglycans to form the expanded HA-rich oocyte-cumulus extracellular matrix (ECM), where the heavy chains of the serum derived inter-α-trypsin inhibitor family (IαI) bind covalently to HA. No study has been performed on the occurrence and regulation of this process during oocyte maturation in species other than mouse and pig, although, the heavy chains (of IαI)-HA complex was purified from human amniotic membrane. The present review pointing out that: 1/ formation of expanded HA-rich oocyte-cumulus ECM is dependent on the presence of IαI molecules, 2/ the heavy chains of IαI molecules identified in the serum are covalently linked to HA during cumulus expansion in mouse and pig, 3/ the family of IαI molecules can freely cross the blood-follicle barrier, and the follicular fluid collected at any stage of folliculogenesis can be successfully used instead of serum to form expanded cumulus ECM in pig, and 4/ proteins of the IαI family can affect reproductive process by modulating the expression of a large number of cellular genes during a preovulatory period. Finally, this review provides clear evidence that IαI family members present in the serum or follicular fluid become responsible for cumulus expansion, as without these proteins, expanded cumulus HA-rich ECM is not formed and HA is released into medium.


Subject(s)
Alpha-Globulins/physiology , Cumulus Cells/cytology , Cumulus Cells/physiology , Extracellular Matrix/physiology , Follicular Phase/physiology , Ovarian Follicle/cytology , Alpha-Globulins/pharmacology , Animals , Cell Proliferation/drug effects , Female , Follicular Fluid/metabolism , Humans , Mice , Ovarian Follicle/physiology , Swine
6.
Domest Anim Endocrinol ; 48: 7-14, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24906923

ABSTRACT

This study was designed to determine whether inhibition of either cyclooxygenase-2 (COX-2) by indomethacin or progesterone receptor (PR) by PR antagonist, RU486, affects oocyte maturation, progesterone production, and covalent binding between hyaluronan (HA) and heavy chains of inter-α trypsin inhibitor, as well as expression of cumulus expansion-associated proteins (HA-binding protein, tumor necrosis factor α-induced protein 6, pentraxin 3) in oocyte-cumulus complexes (OCCs). The experiments were based on freshly isolated porcine OCC cultures in which the consequences of PR and COX-2 inhibition on the final processes of oocyte maturation were determined. Granulosa cells (GCs) and OCCs were cultured in medium supplemented with FSH/LH (both 100 ng/mL) in the presence/absence of RU486 or indomethacin. Western blot analysis, (3)H-glucosamine hydrochloride assay, immunofluorescence, and radioimmunoassay were performed. Only treatment with RU486 (25 µM) caused a decrease in the number of oocytes that reached germinal vesicle breakdown and metaphase II stage compared with indomethacin (100 µM) or FSH/LH treatment alone after 44 h. All treated OCCs synthesized an almost equal amount of HA. Heavy chains (of inter-α trypsin inhibitor)-HA covalent complexes were formed during in vitro FSH/LH-stimulated expansion in RU486- or indomethacin-treated OCCs. Follicle-stimulating hormone/LH-induced progesterone production by OCCs was increased in the presence of RU486 after 44 h. In contrast, a decrease of FSH/LH-stimulated progesterone production by GCs was detected in the presence of either RU486 or indomethacin after 72 h. We suggest that the PR-dependent pathway may be involved in the regulation of oocyte maturation. Both PR and COX-2 regulate FSH/LH-stimulated progesterone production by OCCs and GCs.


Subject(s)
Cumulus Cells/drug effects , Indomethacin/pharmacology , Mifepristone/pharmacology , Oocytes/drug effects , Progesterone/metabolism , Swine , Animals , C-Reactive Protein/genetics , C-Reactive Protein/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cumulus Cells/physiology , Cyclooxygenase Inhibitors/pharmacology , Extracellular Matrix/metabolism , Follicle Stimulating Hormone , Gene Expression Regulation/drug effects , Hormone Antagonists/pharmacology , Hyaluronic Acid , In Vitro Oocyte Maturation Techniques/veterinary , Luteinizing Hormone , Oocytes/physiology , Serum Amyloid P-Component/genetics , Serum Amyloid P-Component/metabolism
7.
Endocr Regul ; 46(4): 225-35, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23127506

ABSTRACT

UNLABELLED: This review deals with molecular mechanisms controlling three important ovarian follicular processes: 1) expansion of the cumulus, 2) synthesis of the hyaluronan (HA), and 3) production of the progesterone in oocyte cumulus complexes (OCCs). The expansion of the mice cumuli induced by FSH or 8-bromo cAMP is dependent upon a specific factor(s) secreted by the oocyte (called "cumulus expansion enabling factor", CEEF). The porcine oocytes produce at least two factors that have influence on the formation and stability of the preovulatory extracellular cumulus matrix (ECM), although oocytectomy does not alter the ability of the cumulus cells to respond to FSH and forskolin by increased cAMP content, HA synthesis, and subsequent cumulus expansion, The net synthesis of HA, during the FSH-stimulated expansion of the OCCs in the presence of serum, correlates directly with accumulation of glycosaminoglycans in the ECM. In pig, insulin growth factor 1 (IGF1) is a component of the serum that promotes the FSH-stimulated synthesis and retention of HA within the expanded ECM by phosphatidylinositol 3-kinase (PI3K)/v-akt murine thymoma viral oncogene homolog (AKT)- and mitogen-activated kinase 3 and 1 (MAPK3/1)-dependent mechanisms. Mouse, porcine, bovine, and rat oocytes produce CEEF(s). Possible candidate for the CEEF in the mouse is growth differentiation factor 9 (GDF9) secreted by oocytes. In pig, GDF9 mRNA is expressed not only in the oocytes but also in the cumulus and mural granulosa cells of the growing and preovulatory follicles, although the relative abundance of the GDF9 in the somatic cells is approximately 4 times lower than in the oocytes. Cross talk between FSH/ epidermal growth factor receptor (EGFR) and transforming growth factor ß (TGFß)/GDF9 signaling pathways is essential for functional activities of the porcine OCCs, since FSH enhances EGF-induced tyrosine phosphorylation of EGFR, indicating that FSH signaling pathway may stimulate specific EGFR-regulating proteins. Also, FSH-induced synthesis of both HA and progesterone is reduced but not abolished by AG1478 (EGFR tyrosine kinase inhibitor), indicating that other signaling pathways elicited by FSH are operating in parallel. Furthermore, SMAD2/3 signaling pathway is involved in the control of both cumulus expansion and steroidogenesis in porcine OCCs, since SMAD2/3 activation by GDF9/ TGFß produced by oocyte and/or cumulus cells, significantly affects gonadotropin-induced HA and progesterone synthesis by porcine cumulus cells. KEYWORDS: oocyte-cumulus complex, hyaluronan, progesterone, cumulus expansion.


Subject(s)
Cumulus Cells/physiology , Hyaluronic Acid/biosynthesis , In Vitro Oocyte Maturation Techniques , Oocytes/physiology , Swine , Animals , Cattle , Cell Proliferation , Cumulus Cells/metabolism , Female , In Vitro Oocyte Maturation Techniques/methods , In Vitro Oocyte Maturation Techniques/veterinary , Mice , Models, Biological , Oocytes/metabolism , Rats , Swine/metabolism , Swine/physiology
8.
Domest Anim Endocrinol ; 42(1): 50-62, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22032857

ABSTRACT

Porcine oocyte-cumulus complexes (OCCs) form an expanded cumulus extracellular matrix (ECM) in response to gonadotropins during meiotic maturation. Essential components of ECM are hyaluronan (HA), tumor necrosis factor α-induced protein 6 (TNFAIP6) and heavy chains (HC) of interalpha-trypsin inhibitor. To form expanded cumulus ECM, intermediate complexes (TNFAIP6-HC) must bind to HA to allow HC transfer onto HA. Protein turnover by the ubiquitin-proteasome pathway is poorly characterized in this process. It is known that the specific proteasomal inhibitor MG132 prevents cumulus expansion and formation of ECM. To determine whether inhibition of proteasomal proteolysis with MG132 affects cumulus cell steroidogenesis and expression of the cumulus expansion-related components (hyaluronan synthase type 2, HAS2, TNFAIP6) we cultured porcine OCCs and granulosa cells (GCs) in a medium supplemented with FSH/LH. Methods performed included real-time reverse transcription PCR, immunofluorescence and RIAs. The expression of TNFAIP6 and HAS2 transcripts increased significantly after the stimulation of OCCs and GCs with FSH/LH. In contrast, treatment with MG132 reduced the expression of TNFAIP6 and HAS2. Hyaluronan was detected with biotinylated HA-binding proteins within FSH/LH-stimulated expanded OCCs but not in those treated with MG132. Progesterone production, although increased almost three times after OCCs stimulation with FSH/LH, was significantly suppressed by MG132. The FSH/LH-stimulated a 40-fold increase in progesterone secretion by GCs was inhibited in the presence of MG132. In conclusion, MG132 affects progesterone secretion and expression of cumulus expansion-related components by cumulus and GCs, suggesting the requirement of ubiquitin-proteasome pathway-regulated protein turnover for formation of ECM during cumulus expansion in the preovulatory period in the pig.


Subject(s)
Cumulus Cells/metabolism , Extracellular Matrix/metabolism , Oocytes/metabolism , Progesterone/biosynthesis , Proteasome Inhibitors , Animals , Cell Adhesion Molecules/biosynthesis , Cumulus Cells/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Extracellular Matrix/drug effects , Female , Leupeptins/pharmacology , Oocytes/drug effects , Proteasome Endopeptidase Complex/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction/veterinary , Swine
9.
Cent Eur J Public Health ; 12 Suppl: S62-4, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15141982

ABSTRACT

Studies aimed at the influence of smoking on reproductive functions have found out fertility disorders in smokers occurring at any stage of reproductive processes. In our experiments the role of cadmium, nicotine and anabasine was investigated in the expansion of oocyte-cumulus complexes (OCC) isolated from large antral porcine follicles. Suppression of FSH-induced cumulus expansion and significant inhibition of synthesis and accumulation of hyaluronic acid in the cell/matrix compartment of the OCC was observed in the presence of different concentrations of tested compounds. The suppressive effect of cadmium and tobacco alkaloids on the cumulus expansion was accompanied by decreased progesterone production by cumulus cells during 42 h incubation of the OCC with FSH.


Subject(s)
Alkaloids/pharmacology , Anabasine/pharmacology , Cadmium/pharmacology , Granulosa Cells/drug effects , Hyaluronic Acid/biosynthesis , Nicotine/pharmacology , Ovarian Follicle/drug effects , Animals , Female , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/metabolism , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Swine
10.
Reprod Suppl ; 61: 55-70, 2003.
Article in English | MEDLINE | ID: mdl-14635927

ABSTRACT

Oocytes secrete factors that control cumulus and granulosa functions, including cumulus expansion and steroid hormone production. Some members of the transforming growth factor beta (TGFbeta) superfamily influence these activities, yet it is still not determined conclusively whether any of these superfamily members are the previously reported oocyte-secreted factors. The aim of this study was to examine the effects of TGFbeta1 and growth differentiation factor 9 (GDF-9) on cumulus expansion and progesterone production by mouse oocytectomized (OOX) complexes in culture. TGFbeta1 mimics the effects of oocytes by both enabling cumulus expansion and inhibiting progesterone production; however, neutralizing antibodies to TGFbeta1 in cultures of cumulus-oocyte complexes (COCs) or in co-cultures of OOX complexes failed to inhibit the ability of oocytes to enable cumulus expansion or inhibit progesterone production. Activin A had no effect on progesterone production by OOX complexes. In experiments using oocytes obtained from mice with deficient expression of GDF-9, OOX complexes cultured in the presence of heterozygous oocytes were capable of full expansion, whereas OOX complexes cultured with oocytes from GDF-9 null mice did not expand. Similarly, GDF-9 null oocytes failed to suppress FSH-induced progesterone production by OOX complexes. These results support the hypothesis that GDF-9 is the cumulus expansion enabling factor produced by mouse oocytes and that GDF-9 also inhibits cumulus progesterone production; however, the possibility remains that loss of GDF-9 may indirectly affect the ability of oocytes to produce the factors that regulate cumulus cell activity.


Subject(s)
Oocytes/physiology , Progesterone/biosynthesis , Transforming Growth Factor beta/pharmacology , Zona Pellucida/metabolism , Activins/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Bone Morphogenetic Protein 15 , Coculture Techniques , Female , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/metabolism , Growth Differentiation Factor 9 , Inhibin-beta Subunits/pharmacology , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Biological , Transforming Growth Factor beta/immunology , Zona Pellucida/drug effects
11.
Physiol Res ; 52(3): 383-7, 2003.
Article in English | MEDLINE | ID: mdl-12790772

ABSTRACT

The role of alkaloids in cigarette smoke was investigated in the cumulus expansion of oocyte-cumulus complexes (OCC) isolated from large antral porcine follicles. Suppression of the cumulus expansion stimulated by FSH was observed in the presence of different concentration of cadmium, anabasine and nicotine but not its metabolite cotinine. There were comparable inhibitory effects of cadmium and nicotine on the synthesis and accumulation of hyaluronic acid in the cell/matrix compartment of OCC. The inhibitory effect of tested compounds on the cumulus expansion was accompanied by decreased progesterone synthesis by cumulus cells during 42 h incubation of OCC with FSH. The results suggest that cigarette smoking may affect intrafollicular processes, which are responsible for normal ovulation and fertilization.


Subject(s)
Alkaloids/pharmacology , Granulosa Cells/drug effects , Hyaluronic Acid/biosynthesis , Ovarian Follicle/drug effects , Progesterone/biosynthesis , Anabasine/pharmacology , Animals , Cadmium/pharmacology , Cotinine/pharmacology , Female , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/metabolism , Nicotine/pharmacology , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Progesterone/metabolism , Swine , Nicotiana/chemistry
12.
Anim Reprod Sci ; 65(1-2): 115-26, 2001 Jan 31.
Article in English | MEDLINE | ID: mdl-11182513

ABSTRACT

The role of granulosa cell conditioned media (CM) containing luteinization stimulator (LS), and the role of EGF in the cumulus expansion of oocyte-cumulus complexes (OCC) isolated from large antral follicles was investigated. The CM were prepared by incubation of granulosa cells isolated from large antral follicles. After 24h incubation, more than 61 or 64% of OCC expanded to the +3 and +4 stage in the presence of CM (50%) or EGF (10ng/ml), respectively. The stimulatory effect of LS and EGF on the cumulus expansion was accompanied by the enhanced hyaluronic acid synthesis. Complete suppression of cumulus expansion stimulated by LS and EGF was observed in the presence of 10 micromol/l genistein (tyrosine kinase inhibitor), in the presence of 10mmol/l LiCl (the inhibitor of inositol 1,4,5-trisphosphate metabolism), and 100 micromol/l gallopamil, verapamil and norverapamil (calcium channel blockers). Stimulatory effect of EGF on the cumulus expansion of OCC isolated from large follicles was accompanied by the increased cumulus cell progesterone production. However, EGF did not affect the progesterone production by OCC isolated from small follicles. To determine whether EGF could modulate the granulosa cell steroidogenesis also, the effect of EGF on granulosa cells isolated from large (LGC) and small (SGC) follicles was compared. EGF (10ng/ml) failed to affect the progesterone synthesis during 72h culture of SGC but significantly enhanced the LGC progesterone production. Our results indicate that luteinization factor stimulates the cumulus expansion and hyaluronic acid synthesis by the OCC isolated from large antral follicles. The mechanism of LS- and EGF-induced cumulus expansion may involve tyrosine kinase activation and calcium mobilization. In addition, these results indicate the different response of porcine cumulus and granulosa cells originating from small and large follicles on the stimulatory effect of EGF.


Subject(s)
Biological Factors/physiology , Granulosa Cells/physiology , Ovarian Follicle/physiology , Progesterone/biosynthesis , Swine/physiology , Animals , Calcium Channel Blockers/pharmacology , Cell Division/physiology , Culture Media, Conditioned , Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/physiology , Female , Follicle Stimulating Hormone/physiology , Genistein/pharmacology , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Growth Inhibitors/pharmacology , Hyaluronic Acid/biosynthesis , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Progesterone/metabolism , Radioimmunoassay/veterinary , Signal Transduction/physiology
13.
Physiol Res ; 50(6): 627-30, 2001.
Article in English | MEDLINE | ID: mdl-11829325

ABSTRACT

The role of gossypol in the cumulus expansion of oocyte-cumulus complexes (OCC) isolated from large antral porcine follicles was investigated. Marked suppression of cumulus expansion stimulated with follicle-stimulating hormone (FSH) and epidermal growth factor (EGF) was observed in the presence of different concentrations of gossypol. Comparable inhibitory effects were obtained in the presence of NO donor, S-nitroso-N-acetylpenicillamine or sodium nitroprusside, suggesting that the inhibitory effect of gossypol may be mediated via NO generation. The inhibitory effect of gossypol on cumulus expansion of OCC was accompanied by inhibition of progesterone secretion of OCC and the decrease of [125I]EGF binding to granulosa cells.


Subject(s)
Epidermal Growth Factor/pharmacology , Follicle Stimulating Hormone/pharmacology , Follicular Phase/drug effects , Gossypol/pharmacology , Oocytes/drug effects , Animals , Epidermal Growth Factor/metabolism , Female , In Vitro Techniques , Iodine Radioisotopes , Oocytes/cytology , Oocytes/metabolism , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Progesterone/metabolism , Swine
14.
Biol Reprod ; 63(4): 1149-56, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10993839

ABSTRACT

To demonstrate secretion of cumulus expansion-enabling factor (CEEF) by porcine oocytes, we used an interspecies testing system. Porcine oocytes were used to condition culture medium, and the presence of CEEF was tested using mouse oocytectomized complexes (OOX), which require CEEF for expansion. Follicle-stimulating hormone-stimulated expansion and synthesis of hyaluronic acid (HA) by mouse OOX were assessed after 18 h of culture in media conditioned by porcine oocytes: 1) at different stages of maturation and 2) in which maturation was inhibited with a specific inhibitor of cdk-kinases, butyrolactone I. Fully grown (GV-germinal vesicle), late-diakinesis (LD), metaphase I (MI), and metaphase II (MII) oocytes were prepared by culture of oocyte-cumulus complexes (OCC) for 0, 22, 27, and 42 h, respectively. To block GV breakdown, porcine oocytes were cultured for 27 h in medium supplemented with butyrolactone I (50 microM). Medium conditioned by oocytes in GV, LD, and after butyrolactone I block allowed full expansion of >90% of mouse OOX, whereas oocytes in MI and MII caused disintegration of mouse OOX without cumulus mucification. To measure synthesis of HA by cumulus cells, 25 mouse OOX were cultured in the conditioned media in the presence of 2.5 microCi of D-[6-(3)H]glucosamine hydrochloride. After 18 h, incorporation of the [(3)H]glucosamine into HA was determined either in complexes (retained HA) or in medium plus complexes (total HA). Total HA accumulation by mouse OOX was not different from that of intact OCC. However, oocytes in GV, LD, and after butyrolactone I treatment enabled mouse OOX to retain significantly more HA within the complex than oocytes in MI and MII. The results indicate that secretion of factors that promote the retention of HA within the complex is developmentally regulated during oocyte maturation.


Subject(s)
Oocytes/cytology , Oocytes/physiology , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/pharmacology , Animals , Cells, Cultured , Culture Media, Conditioned , Cyclin B/drug effects , Cyclin B/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Female , Follicle Stimulating Hormone/pharmacology , Histones/drug effects , Histones/metabolism , Hyaluronic Acid/biosynthesis , Meiosis , Mice , Mice, Inbred ICR , Oocytes/drug effects , Ovarian Follicle/cytology , Paracrine Communication , Swine
15.
Mol Reprod Dev ; 56(1): 63-73, 2000 May.
Article in English | MEDLINE | ID: mdl-10737968

ABSTRACT

Epidermal growth factor (EGF) efficiently stimulates expansion of mouse and rat oocyte-cumulus complexes (OCC). Contradictory data have been published by several laboratories about the ability of EGF to stimulate expansion of porcine OCC. We assumed that these contradictions may have resulted from heterogeneous conditions used for isolation, culture, and assessment of OCC. The present experiments were designed to test the hypothesis that porcine OCC acquire the ability to synthesize hyaluronic acid (HA) and undergo expansion following EGF-stimulation gradually during the growth of follicles. For this reason, we isolated OCC from follicles of different sizes and assessed quantity of produced HA and proportions of expanding OCC after stimulation by EGF. In addition, we assessed in those OCC changes in morphology of cumulus cells and assembly of F-actin microfilaments, which are necessary for expansion to occur. Finally, nuclear maturation of EGF-stimulated OCC was assessed and its relationship with occurrence of expansion was evaluated. In all experiments, OCC stimulated with FSH were used as positive controls. The results showed that EGF did not stimulate production of HA, rearrangement of F-actin and expansion in OCC isolated from small follicles (<4 mm in diameter). OCC isolated from large preovulatory follicles (6-7 mm in diameter and PMSG-stimulated follicles) underwent efficient expansion when stimulated by EGF (93% and 100%, respectively). EGF dramatically stimulated total production of HA in these OCC and its retention in extracellular matrix of the expanding cumulus. Cumulus cells of the large OCC underwent essential changes of their morphology and extensive rearrangement of F-actin microfilaments following stimulation with EGF. Interestingly, EGF enhanced nuclear maturation of OCC isolated from both small and large follicles, which suggest diversity of signaling pathways controlling maturation and expansion. FSH caused cumulus expansion, F-actin remodeling, and enhancement of oocyte nuclear maturation in OCC originated from both small and large follicles. We conclude that EGF can stimulate expansion of porcine OCC in vitro; however, only of those isolated from large follicles. This indicates that EGF may have a physiological role in regulation of porcine cumulus expansion in preovulatory follicles, presumably as a mediator of signals elicited by the LH surge.


Subject(s)
Actins/metabolism , Epidermal Growth Factor/pharmacology , Oocytes/drug effects , Animals , Cell Division/drug effects , Cell Nucleus/metabolism , Cell Nucleus/physiology , Cells, Cultured , Female , Follicle Stimulating Hormone/pharmacology , Hyaluronic Acid/metabolism , Oocytes/metabolism , Oocytes/physiology , Ovarian Follicle/cytology , Swine
16.
Biol Reprod ; 61(3): 569-74, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10456830

ABSTRACT

Mouse oocytes secrete a factor that enables cumulus cells to undergo expansion in response to FSH (1 microg/ml), whereas expansion of the porcine cumulus oophorus has been shown to be independent of the oocyte. The aim of this study was to assess FSH-induced synthesis of hyaluronic acid (HA) by porcine cumulus cells before and after oocytectomy. In addition, we studied the effect of insulin-like growth factor-I (IGF-I) on the ability of cumulus cells to synthesize and retain HA in response to FSH in serum-free medium. Porcine oocyte-cumulus complexes and complexes from which the oocytes had been removed by oocytectomy were cultured for 24 h in the presence of 2.5 microCi of D-[6-(3)H]glucosamine hydrochloride, fetal calf serum (FCS, 5%), and FSH. After 24 h, incorporation of [(3)H]glucosamine into HA was measured either in complexes alone (retained HA) or in medium plus complexes (total HA). Specificity of incorporation of radioactivity into HA was confirmed by the sensitivity to highly specific Streptomyces hyaluronidase. Our results suggest that 1) the synthesis of HA by pig cumulus cells in vitro is stimulated by FSH and that oocytectomy does not change this synthesis; 2) oocytes do not influence retention of HA within the complex; 3) FSH-induced synthesis of HA by cumulus cells is decreased in medium with polyvinylpyrrolidone (PVP)-supplemented (total and retained HA) compared to FCS-supplemented medium; 4) IGF-I enabled cumulus cells to synthesize HA in response to FSH in PVP-supplemented medium in a manner similar to that observed when serum is present in the medium.


Subject(s)
Hyaluronic Acid/biosynthesis , Insulin-Like Growth Factor I/pharmacology , Oocytes/physiology , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Swine , Animals , Cells, Cultured , Culture Media, Serum-Free , Female , Follicle Stimulating Hormone/pharmacology
17.
Mol Reprod Dev ; 49(2): 141-9, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9444657

ABSTRACT

The objective of this study was to find out whether porcine cumulus and mural granulosa cells can secrete cumulus expansion-enabling factor (CEEF). Culture drops of M-199 medium were conditioned with denuded porcine oocytes (1 oocyte/microliter), cumulus cells from oocytectomized complexes (1 OOX/microliter), pieces of mural granulosa isolated from preantral to preovulatory follicles (1000 cells/microliter), or oviductal cells (1000 cells/microliter) for 24 hr. The production of CEEF was assessed by the addition of mouse OOX and follicle-stimulating hormone (FSH) (1 microgram/ml) to microdrops of the conditioned medium. After 16-18 hr, expansion of the mouse OOX was scored on a scale of 0 to 4 by morphologic criteria. Mouse OOX did not expand in nonconditioned FSH-supplemented medium. Immature porcine oocytes produced +3 to +4 expansion of the mouse OOX. Granulosa cells isolated from preantral and early antral follicles and cumulus cells isolated from all states of follicle development constitutively secreted CEEF under in vitro conditions. Mural granulosa cells of small, medium, and preovulatory (PMSG) follicles also secreted CEEF in vitro; however, FSH or leutenizing hormone (LH) stimulation was essential for this secretion. Hormonally induced secretion of CEEF was accompanied by expansion of the mural granulosa itself. Granulosa cells isolated from follicles of gilts 20 hr after PMSG and human chorionic gonadotropin (hCG) administration did not produce CEEF and did not expand in response to FSH and LH in vitro. CEEF activity also was found in the follicular fluid of small antral follicles, was reduced in medium follicles, and was not detectable in PMSG-stimulated follicles. However, CEEF activity was reestablished in the follicular fluid of preovulatory follicles by hCG injection, conceivably due to increased production of CEEF by cumulus cells. We conclude that (1) porcine cumulus and mural granulosa cells are capable of CEEF production in vitro and (2) autocrine secretion of CEEF by cumulus cells is involved in regulation of porcine cumulus expansion both in vitro and in vivo.


Subject(s)
Growth Substances/metabolism , Oocytes/metabolism , Ovarian Follicle/metabolism , Animals , Cell Communication/physiology , Cell Division/physiology , Cell Separation , Cells, Cultured , Culture Media, Conditioned , Fallopian Tubes/metabolism , Female , Granulosa Cells/metabolism , Granulosa Cells/physiology , Humans , Mice , Oocytes/cytology , Oocytes/physiology , Ovarian Follicle/cytology , Swine
18.
Zygote ; 6(4): 299-309, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9921640

ABSTRACT

The meiotic competence and meiosis resumption of Blue fox (Alopex lagopus) oocytes from anoestrous animals were followed. Oocyte-cumulus complexes (OCC) were cultured in modified TC 199 medium with or without FSH, recombinant bovine somatotrophin (bST) and okadaic acid (OA). The results showed that oocytes less than 100 microns in diameter did not achieve germinal vesicle breakdown (GFBD) by 72 h of culture, which indicates their meiotic incompetence. Oocytes larger than 100 microns in diameter underwent GVBD after 48 h of culture (27%) and reached metaphase II (MII) after 72 and 96 h (20% and 27%) in control medium. Both bST and OA accelerated resumption of meiosis (bST: 55% GVBD and 42% MII after 48 h; OA: 66% GVBD after 18 h). In contrast, FSH significantly reduced meiosis resumption (only 3% GVBD and MII after 72 h) and induced changes in the shape of cumulus granulosa (CG) cells and F-actin assembly typical for cumulus expansion. However, the innermost layers of CG cells (corona radiata) remained connected with the oocyte via gap junctions until the end of culture. Cumuli of oocytes cultured in control, bST-supplemented or OA-supplemented medium did not expand (changes in cell shape and F-actin redistribution did not occur). Moreover, especially in media with bST and OA an increased detachment and rapid disconnection of their gap junctions with the oocyte were observed. These results suggest that under in vitro conditions FSH stimulates expansion of the CG cells and the attached membrana granulosa cells but in contrast it secures heterologous gap junctions between cytoplasmic processes of the corona radiata cells and oolemma during 3 days of culture. Thus, in agreement with the in vivo situation in which Canidae oocytes are ovulated in the GV stage, the cumulus, mainly corona radiata cells, controls resumption of meiosis in Blue fox oocytes under in vitro conditions also.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Foxes/growth & development , Growth Hormone/pharmacology , Okadaic Acid/pharmacology , Oocytes/drug effects , Oocytes/growth & development , Animals , Cattle , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Female , Foxes/anatomy & histology , Foxes/physiology , Granulosa Cells/physiology , In Vitro Techniques , Meiosis/drug effects , Meiosis/physiology , Microscopy, Electron , Oocytes/physiology
19.
Exp Cell Res ; 236(1): 193-200, 1997 Oct 10.
Article in English | MEDLINE | ID: mdl-9344599

ABSTRACT

The meiosis resumption process has been related to spontaneous cytoplasmic InsP3-dependent calcium oscillations in fully grown mouse oocytes. Our purpose was to determine whether the acquisition of meiotic competence during the growth phase of oogenesis was associated with that of Ca2+ oscillations and whether these oscillations were dependent on the phosphoinositide cycle. We used confocal laser scanning microscopy to image free calcium ions in fluo-3/AM-loaded oocytes recovered from 12- to 26-day-old mice for 15 min following follicular release. As expected, oocytes isolated from 12-day-old mice were totally incompetent to undergo GVB in vitro, whereas the GVB rate increased progressively with mouse age and oocyte diameter. The percentage of oocytes exhibiting spontaneous calcium oscillations and that of oocytes resuming meiosis were similarly correlated with the female age, with incompetent oocytes failing to exhibit spontaneous Ca2+ oscillations. It is noteworthy that regardless of the stage of growth, thapsigargin induced an ooplasmic calcium release from the InsP3-sensitive stores when it was added to the culture medium. However, intracytoplasmic microinjection of InsP3 induced a shorter sequence of Ca2+ oscillations in 12-day-old mouse oocytes than in 15-day-old mouse oocytes and, whereas InsP3 increased the GVB rate at 15 days, it was unable to induce GVB at 12 days. These data lead us to conclude that the acquisition of meiotic competence is related to the functionality of the InsP3 pathway and, correspondingly, to the oocyte's ability to generate spontaneous cytoplasmic InsP3-dependent calcium oscillations.


Subject(s)
Calcium/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Meiosis/physiology , Oocytes/cytology , Signal Transduction/physiology , Age Factors , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Female , Inositol 1,4,5-Trisphosphate/pharmacology , Mice , Mice, Inbred Strains , Microinjections , Oocytes/drug effects , Oocytes/physiology , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Periodicity , Thapsigargin/pharmacology
20.
Reprod Nutr Dev ; 33(5): 419-28, 1993.
Article in English | MEDLINE | ID: mdl-8142026

ABSTRACT

Resumption of meiosis was inhibited in mouse oocyte cumulus complexes (OCC) co-cultured with pig membrane granulosa (PMG). After 3 and 6 h of co-culture these oocytes possessed an intact nuclear envelope and their nucleolar surface was associated with granules approximately 80 nm in diameter. Preincubation of OCCs for 30, 45, 60 or 90 min followed by co-culture with PMG for 2 h of either OCCs or denuded oocytes resulted in germinal vesicle breakdown (GVBD) in approximately 0, 30, 70 and 100% mouse OCCs and in approximately 30, 60, 80 and 100% denuded oocytes, respectively. It seems that the inhibitory contact between mouse oocytes and PMG was established during the first h of co-culture. After isolation from antral follicles the oocytes contained 2.75 fmol cyclic adenosine 3', 5'-monophosphate (cAMP). When OCCs were co-cultured for 1, 2 or 3 h with PMG, the amount of cAMP per oocyte was 1.34, 1.33 and 1.51 fmol, respectively. After culture of OCCs in control medium the amount of cAMP was 1.21, 1.39 and 2.16 fmol, respectively. The present results suggest that the inhibitory activity of PMG is not species-specific. Moreover, PMG prevented resumption of meiosis in mouse oocytes in spite of the cAMP drop in oocyte cytoplasm characteristic of the resumption of meiosis.


Subject(s)
Cyclic AMP/physiology , Meiosis/physiology , Oocytes/physiology , Animals , Basement Membrane , Cells, Cultured , Cyclic AMP/pharmacology , Female , Granulosa Cells/physiology , Meiosis/drug effects , Mice , Microscopy, Electron , Oocytes/drug effects , Oocytes/ultrastructure , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...