Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Toxicol Appl Pharmacol ; 473: 116599, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37328116

RESUMO

Studies have indicated that glyphosate induces endocrine disruption and may adversely affect the male reproductive system. However, evidence of its effects on ovarian function is poorly understood so far, making further studies necessary on the mechanisms of the glyphosate toxicity in the female reproductive system. The aim of this work was to evaluate the effect of a subacute exposure (28 days) to the glyphosate-based formulation Roundup® (1.05, 10.5 and 105 µg/kg b.w. of glyphosate) on steroidogenesis, oxidative stress, systems involved in cell redox control and histopathological parameters in rat ovaries. Hence we quantify plasma estradiol and progesterone by chemiluminescence; non-protein thiol levels, TBARS, superoxide dismutase and catalase activity by spectrophotometry; gene expression of steroidogenic enzymes and redox systems by real-time PCR; and ovarian follicles by optical microscopy. Our results demonstrated that oral exposure increased progesterone levels and the mRNA expression of 3ß-hydroxysteroid dehydrogenase. Histopathological analysis revealed a decrease in the number of primary follicles and an increase in the number of corpus luteum in rats exposed to Roundup®. An imbalance of the oxidative status was also evidenced by decreasing the catalase activity at all groups exposed to the herbicide. Increased lipid peroxidation and gene expression of glutarredoxin and decreased of glutathione reductase were also observed. Our results indicate that Roundup® causes endocrine disruption of hormones related to female fertility and reproduction and changes the oxidative status by altering antioxidant activity, inducing lipid peroxidation, as well as changing the gene expression of the glutathione-glutarredoxin system in rat ovaries.


Assuntos
Herbicidas , Ovário , Ratos , Masculino , Feminino , Animais , Progesterona , Catalase/genética , Catalase/metabolismo , Herbicidas/toxicidade , Glutarredoxinas/farmacologia , Antioxidantes/farmacologia , Glutationa/metabolismo , Estradiol/farmacologia , Expressão Gênica
2.
Reprod Biol ; 23(1): 100709, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36403532

RESUMO

Cumulus cells from cumulus-oocyte complexes (COC) matured in vitro in serum-free medium show high incidence of apoptosis and DNA double-strand breaks (DSB). This study aimed to characterize the transcript expression profile of selected genes involved in DNA repair mechanisms in bovine cumulus cells cultured with bovine serum albumin (BSA) or fetal calf serum (FCS). Briefly, bovine cumulus-oocyte complexes were in vitro matured with either, 0.4% BSA or 10% FCS for 3, 6, 12 or 24 h. The total RNA of cumulus cells was used for real-time PCR analysis. Transcript abundance of XRCC6, XRCC5, DNAPK, GAAD45B, TP53BP1, RAD50, RAD52, ATM and BRCA2 target genes changed as the IVM proceeded (P < 0.05). However, an interaction between protein source (FCS or BSA) and time was not detected (P ≥ 0.05). Cumulus cells from COCs matured with BSA presented higher mRNA expression of two genes compared to FCS group: TP53BP1 at 6 h and BRCA1 at 3, 6, 12 and 24 h (P < 0.05). In summary, our results showed for the first time the expression profile of the key genes involved in DSB repair mechanisms in cumulus cells obtained from bovine COCs matured with FCS or BSA. The higher mRNA expression of BRCA1 and TP53BP1 and lower mRNA expression of TNFAIP6 suggests an increase in apoptosis rate and DNA damage in cumulus cells cultured in BSA-supplemented medium and may explain, at least to some extent, the reduced developmental potential of bovine oocytes matured in serum-free medium.


Assuntos
Células do Cúmulo , Soroalbumina Bovina , Feminino , Animais , Células do Cúmulo/metabolismo , Oócitos/metabolismo , Reparo do DNA , RNA Mensageiro/metabolismo , Células Cultivadas
3.
Mol Cell Endocrinol ; 541: 111524, 2022 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-34856345

RESUMO

Failure to ovulate is a major cause of infertility. The critical pathway that induces ovulation involves the EGF and MAPK phosphorylation, but studies in rodents have suggested that the Hippo activator, YAP, is also involved. It is unknown whether YAP-dependent transcriptional activity is important for the LH- or EGF-induced ovulatory cascade in monovulatory species such as the cow. Using a well-defined preovulatory GC culture system, we employed pharmacological inhibitors to demonstrate that YAP signaling is critical for expression of EGFR and downstream target genes EREG, EGR1 and TNFAIP6. Most importantly, by using an ultrasound guided follicle injection system, we also showed that the classic Hippo signaling inhibitor Verteporfin inhibits GnRH-induced ovulation in vivo in cattle. In conclusion, YAP transcriptional activity is critical for EGF-like cascade induced by LH to promote ovulation in a monovulatory species.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Células da Granulosa/metabolismo , Ovulação/fisiologia , Proteínas de Sinalização YAP/fisiologia , Animais , Bovinos , Células Cultivadas , Feminino , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/fisiologia , Hormônio Luteinizante/farmacologia , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/metabolismo , Ovulação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas de Sinalização YAP/genética
4.
Zygote ; 30(2): 239-243, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34405788

RESUMO

The Hippo pathway is involved in the proliferation of intrafollicular cells and in early embryonic development, mainly because effectors of this pathway are key transcription regulators of genes such as CTGF and CYR61, which are involved in cell proliferation. Recent studies by our group found that fibroblast growth factor 18 (FGF18) is present in the fallopian tube during early embryonic development, leading to the hypothesis that FGF18 may have a role during embryonic development. Therefore, the aim of the following study was to determine whether FGF18 modulates the expression of Hippo pathway target genes, CTGF and CYR61, during oocyte maturation and early embryonic development. Three experiments were carried out, with in vitro maturation (IVM) of cumulus-oocyte complexes (COCs) and embryo culture. In experiment one, FGF18 (100 ng/ml) induced an increase (P < 0.05) in CTGF gene expression at 12 h post-exposure. In experiment two, FGF18 (100 ng/ml) induced a reduction (P < 0.05) in CTGF expression at 3 h post-exposure. In the third experiment, day 7 embryos exposed to FGF18 during oocyte IVM expressed greater CTGF mRNA abundance, whereas FGF18 exposure during embryo in vitro embryo culture did not alter CTGF expression in comparison with untreated controls. The preliminary data presented here show that FGF18 modulates CTGF expression in critical periods of oocyte nuclear maturation, cumulus expansion and early embryonic development in cattle.


Assuntos
Células do Cúmulo , Técnicas de Maturação in Vitro de Oócitos , Animais , Blastocisto , Bovinos , Desenvolvimento Embrionário , Feminino , Fatores de Crescimento de Fibroblastos , Técnicas de Maturação in Vitro de Oócitos/veterinária , Oócitos , Gravidez , Dados Preliminares , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
5.
J Appl Toxicol ; 41(8): 1180-1187, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33140417

RESUMO

Contamination of animal feed with Fusarium spp results in accumulation of mycotoxins including deoxynivalenol. In animals, deoxynivalenol is metabolized to de-epoxy deoxynivalenol (DOM-1), which is generally considered to be a non-toxic metabolite; however, recent studies demonstrated that DOM-1 can reduce steroid production and induce apoptosis in the bovine ovary. The objectives of this study were to assess the effects of DOM-1 on applied aspects of reproductive function in cattle, specifically sperm function and embryo development in vitro and follicle growth and superovulatory responses in vivo. The effect of naturally contaminated feed on superovulatory responses was assessed; a dose of 6 ppm deoxynivalenol increased blood DOM-1 concentrations to 20 ng/ml, but this did not alter the number of viable embryos recovered on day 7. However, intrafollicular injection of DOM-1 (100 ng/ml) directly into the growing dominant follicle resulted in cessation of follicular growth over the subsequent 3 days. Treatment with DOM-1 reduced motility of bull spermatozoa over a 10-h period in vitro. Addition of DOM-1 to oocytes in vitro during IVM did not alter rates of cumulus expansion and nuclear maturation, but treatment during IVF reduced the rate of blastocyst formation. These data illustrate that DOM-1 is more biologically active than previously thought and negatively impacted reproductive outcomes in cattle.


Assuntos
Desenvolvimento Embrionário/efeitos dos fármacos , Micotoxinas/toxicidade , Motilidade dos Espermatozoides/efeitos dos fármacos , Tricotecenos/toxicidade , Ração Animal/microbiologia , Ração Animal/toxicidade , Animais , Blastocisto/efeitos dos fármacos , Bovinos , Feminino , Contaminação de Alimentos , Fusarium/metabolismo , Masculino , Micotoxinas/sangue , Oócitos/efeitos dos fármacos , Superovulação/efeitos dos fármacos , Tricotecenos/sangue
6.
J Renin Angiotensin Aldosterone Syst ; 16(2): 275-83, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23764714

RESUMO

INTRODUCTION: This study was based on the hypothesis that some components of the angiotensin-(1-7) (Ang-(1-7)) system are differentially expressed during follicular development and can be involved in the follicular health/atresia transition in bovine. MATERIAL AND METHODS: The largest (F1) and second largest follicles (F2) were collected from cows before (Day 2), during (Day 3), or after (Day 4) the expected moment of follicular deviation. In the second experiment, F1 was induced to atresia through intrafollicular injection of fulvestrant (estrogen receptor-antagonist) and, in both experiments, mRNA expression of the Mas receptor, ACE2, NEP, and PEP was evaluated in the granulosa and theca cells. RESULTS: The mRNA expression of Mas receptor was upregulated in the granulosa cells of F2 after the establishment of follicular deviation, while PEP mRNA increased during and after the deviation process. The mRNA expression of ACE2 was upregulated in the granulosa cells of F1 during and after the follicular deviation. The mRNA expression of NEP was not regulated in F1 and F2. Mas receptor expression increased in the F1 induced to atresia. CONCLUSIONS: mRNA for Mas receptor, ACE2, and PEP are differentially expressed in granulosa cells throughout follicular development and the Mas receptor can be involved with the establishment of follicular dominance.


Assuntos
Angiotensina I/metabolismo , Perfilação da Expressão Gênica , Folículo Ovariano/metabolismo , Fragmentos de Peptídeos/metabolismo , Angiotensina I/genética , Enzima de Conversão de Angiotensina 2 , Animais , Bovinos , Estradiol/análogos & derivados , Estradiol/farmacologia , Feminino , Atresia Folicular/efeitos dos fármacos , Atresia Folicular/genética , Fulvestranto , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Neprilisina/genética , Neprilisina/metabolismo , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/enzimologia , Fragmentos de Peptídeos/genética , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/metabolismo , Prolil Oligopeptidases , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Células Tecais/efeitos dos fármacos , Células Tecais/metabolismo
7.
Biol Reprod ; 92(1): 14, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25411391

RESUMO

Although the various members of the fibroblast growth factor (FGF) family are generally mitotic, one member, FGF18, has been shown to increase the rate of apoptosis of ovarian granulosa cells. In the present study, we first determined whether granulosa cells express FGF18 and we then explored the mechanism through which FGF18 increases apoptosis in vitro. Under culture conditions that favored estradiol secretion and CYP19A1 expression, granulosa FGF18 mRNA levels were barely detectable; however, withdrawing gonadotropic support (follicle-stimulating hormone or insulin-like growth factor 1) reduced levels of CYP19A1 mRNA and increased abundance of mRNA encoding the death ligand FASLG and FGF18. Addition of FGF18, but not FGF2, FGF10, or EGF, increased the proportion of apoptotic cells and frequency of caspase 3 activation, and these effects were abrogated by coculture with estradiol. Addition of FGF18 decreased abundance of mRNA encoding the antiapoptotic proteins GADD45B and MDM2, and increased that encoding the proapoptotic protein BBC3; these effects were reversed by coculture with estradiol. The physiological relevance of FGF18 was determined using an in vivo model: injection of FGF18 directly into growing bovine dominant follicles caused cessation of follicle growth by 24 h after injection. Collectively, these data demonstrate that FGF18 is proapoptotic in vivo and may act through a mechanism involving the BBC3-MDM2 pathway.


Assuntos
Bovinos , Fatores de Crescimento de Fibroblastos/genética , Atresia Folicular/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteínas Reguladoras de Apoptose/fisiologia , Bovinos/genética , Bovinos/fisiologia , Células Cultivadas , Feminino , Fatores de Crescimento de Fibroblastos/farmacologia , Regulação da Expressão Gênica , Células da Granulosa/metabolismo , Células da Granulosa/fisiologia , Folículo Ovariano/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
8.
Theriogenology ; 79(8): 1204-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23510789

RESUMO

The objective was to determine the effects of eCG given on the day of, or 2 days before removal of an intravaginal progestin device, on ovarian follicle diameter, luteal volume, serum progesterone (P4) concentrations, and pregnancy per insemination in a fixed-time AI (FTAI) protocol. Lactating, anestrous, multiparous Bos taurus cross beef cows, 40 to 60 days postpartum, were given estradiol benzoate (2 mg im) and a progestin intravaginal device containing 250 mg of medroxyprogesterone acetate on Day 0 and cloprostenol (0.265 mg) on Day 6. Intravaginal devices were removed on Day 8 and GnRH (100 µg im) was given on Day 9, with timed AI 16 hours later. In experiment 1, cows were randomly assigned to receive 400 IU im eCG on Day 6 (eCG6; N = 8) or Day 8 (eCG8; N = 8), or to not receive eCG (control; N = 8). Dominant follicle diameter on Day 9 in the eCG6 group (10.0 ± 0.5 mm) was larger (P < 0.05) than in the eCG8 (8.6 ± 0.2 mm) or control (8.5 ± 0.4 mm) groups. Corpora lutea (CL) in all cows in the control group underwent premature luteolysis within 10 days after ovulation. Luteal volumes and P4 concentrations 10 and 15 days after ovulation were higher (P < 0.05) in the eCG6 group than in the eCG8 group. In experiment 2, the eCG6 (N = 121) and eCG8 (N = 125) protocols were compared in lactating anestrous cows that underwent FTAI. Pregnancy rate was higher (P < 0.05) in the cows that received eCG on Day 6 (27.3%; 33/121) than on Day 8 (16.0%; 20/125). Furthermore, CL volumes and P4 concentrations were higher (P < 0.05) in the eCG6 group (5784.0 ± 857.3 mm(3) and 8.1 ± 1.3 ng/mL, respectively) than in the eCG8 group (3220.9 ± 505.1 mm(3) and 4.5 ± 0.7 ng/mL, respectively). We concluded that eCG given 2 days before progestin removal in this FTAI protocol for anestrous beef cows increased diameter of the dominant follicle, luteal volume, serum P4 concentrations, and pregnancy rates.


Assuntos
Anestro/efeitos dos fármacos , Corpo Lúteo/efeitos dos fármacos , Gonadotropinas Equinas/farmacologia , Inseminação Artificial/métodos , Folículo Ovariano/efeitos dos fármacos , Taxa de Gravidez , Progesterona/sangue , Anestro/sangue , Anestro/fisiologia , Animais , Bovinos , Tamanho Celular/efeitos dos fármacos , Corpo Lúteo/anatomia & histologia , Sincronização do Estro/métodos , Sincronização do Estro/fisiologia , Feminino , Inseminação Artificial/veterinária , Masculino , Tamanho do Órgão/efeitos dos fármacos , Concentração Osmolar , Folículo Ovariano/citologia , Gravidez , Fatores de Tempo
9.
Exp Cell Res ; 318(16): 2049-58, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22691445

RESUMO

This study investigated the expression of genes controlling homologous recombination (HR), and non-homologous end-joining (NHEJ) DNA-repair pathways in bovine embryos of different developmental potential. It also evaluated whether bovine embryos can respond to DNA double-strand breaks (DSBs) induced with ultraviolet irradiation by regulating expression of genes involved in HR and NHEJ repair pathways. Embryos with high, intermediate or low developmental competence were selected based on the cleavage time after in vitro insemination and were removed from in vitro culture before (36 h), during (72 h) and after (96 h) the expected period of embryonic genome activation. All studied genes were expressed before, during and after the genome activation period regardless the developmental competence of the embryos. Higher mRNA expression of 53BP1 and RAD52 was found before genome activation in embryos with low developmental competence. Expression of 53BP1, RAD51 and KU70 was downregulated at 72 h and upregulated at 168 h post-insemination in response to DSBs induced by ultraviolet irradiation. In conclusion, important genes controlling HR and NHEJ DNA-repair pathways are expressed in bovine embryos, however genes participating in these pathways are only regulated after the period of embryo genome activation in response to ultraviolet-induced DSBs.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades/genética , Desenvolvimento Embrionário/fisiologia , Animais , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Bovinos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fertilização In Vitro , Regulação da Expressão Gênica no Desenvolvimento , Recombinação Homóloga , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Autoantígeno Ku , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/genética , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Raios Ultravioleta
10.
Theriogenology ; 77(9): 1779-87, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22365701

RESUMO

Oocyte meiotic resumption is triggered by the ovulatory gonadotropin surge; in cattle, angiotensin II (AngII) and prostaglandins (PG) are key mediators of this gonadotropin-induced event. Here, we tested the hypothesis that progesterone (P(4)) is also involved in oocyte meiotic resumption induced by the gonadotropin surge. In Experiment I, P(4) induced nuclear maturation in a dose-dependent manner using a coculture of follicular hemisections and cumulus-oocyte complexes. In the second experiment, using an in vivo model, an injection of mifepristone (MIFE; P(4) receptor antagonist) at the antrum of preovulatory follicles prevented GnRH-induced oocyte meiotic resumption in vivo. In Experiment III (coculture system similar to that of Experiment I), MIFE prevented stimulatory effects of AngII on resumption of meiosis, but saralasin (AngII receptor antagonist) did not inhibit P(4) actions. In Experiments IV and V, fibroblast growth Factor 10 (FGF10; known to suppress steroidogenesis in granulosa cells), blocked AngII-but not P(4)-induced oocyte meiotic resumption. Therefore, we inferred that AngII is upstream to P(4) in a cascade to induce meiotic resumption. Previously, we had reported that AngII acted throughout the PGs pathway to modulate nuclear progression. In Experiment V, indomethacin inhibited resumption of meiosis induced by P(4), providing further support to the AngII-P(4) sequential effect on meiotic resumption. In conclusion, we inferred that AngII, P(4) and PGs are sequential steps in the same pathway that culminates with bovine oocyte maturation.


Assuntos
Angiotensina II/metabolismo , Bovinos/sangue , Técnicas de Maturação in Vitro de Oócitos/veterinária , Oócitos/fisiologia , Progesterona/metabolismo , Prostaglandinas/metabolismo , Animais , Fator 10 de Crescimento de Fibroblastos/farmacologia , Indometacina/farmacologia , Luteolíticos/farmacologia , Meiose/fisiologia , Mifepristona/farmacologia , Receptores de Progesterona/antagonistas & inibidores
11.
Endocrinology ; 152(12): 4957-65, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22009728

RESUMO

It is generally understood that angiotensin II (AngII) promotes follicle atresia in rats, although recent data suggested that this may not be true in cattle. In this study, we aimed to determine in vivo whether AngII alters follicle development in cattle, using intrafollicular injection of AngII or antagonist into the growing dominant follicle or the second largest subordinate follicle. Injection of saralasin, an AngII antagonist, into the growing dominant follicle inhibited follicular growth, and this inhibitory effect was overcome by systemic FSH supplementation. Injection of AngII into the dominant follicle did not affect follicular growth, whereas injection of AngII into the second largest follicle prevented the expected atresia of this subordinate follicle, and the treated follicle grew at the same rate as the dominant follicle for the next 24 h. Inhibition of AngII action in the dominant follicle decreased estradiol concentrations in follicular fluid and the abundance of mRNA encoding aromatase, 3ß-hydroxysteroid dehydrogenase, LH receptor, and cyclinD2 in granulosa cells, with minimal effects on theca cells. The effect of AngII on aromatase mRNA levels was confirmed using an in vitro granulosa cell culture system. In conclusion, these data suggest that AngII signaling promotes follicle growth in cattle and does so by regulating genes involved in estradiol secretion and granulosa cell proliferation and differentiation.


Assuntos
Angiotensina II/farmacologia , Folículo Ovariano/crescimento & desenvolvimento , Transdução de Sinais , Angiotensina II/administração & dosagem , Angiotensina II/fisiologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II , Animais , Aromatase , Bovinos , Diferenciação Celular , Proliferação de Células , Estradiol , Feminino , Regulação da Expressão Gênica , Células da Granulosa/citologia , Células da Granulosa/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Saralasina/administração & dosagem , Saralasina/farmacologia , Transdução de Sinais/efeitos dos fármacos
12.
Reproduction ; 136(6): 733-40, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18768665

RESUMO

Angiotensin II (AngII) prevents the inhibitory effect of follicular cells on oocyte maturation, but its involvement in LH-induced meiotic resumption remains unknown. The aim of this study was to assess the involvement of AngII in LH-induced meiotic resumption and of prostaglandins (PGs) in the action of AngII. In the experiment I, seven cows were superovulated, intrafollicularly injected with 10 muM saralasin (a competitive AngII antagonist) or saline when the follicles reached a diameter larger than 12 mm, and challenged with a GnRH agonist to induce an LH surge. Fifteen hours after GnRH, the animals were ovariectomized and the oocytes were recovered to determine the stage of meiosis. The oocytes from follicles that received saline were in germinal vesicle (GV) breakdown (30.8%) or metaphase I (MI; 69.2%) stage while those that received saralasin were in the GV stage (100%; P<0.001) 15 h after GnRH agonist. In another experiment, oocytes were co-cultured with follicular hemisections for 15 h to determine whether PGs mediate the effect of AngII on meiotic resumption. Indomethacin (10 microM) inhibited AngII-induced meiotic resumption (13.4 vs 77.5% MI without indomethacin; P<0.001). Furthermore, the GV oocytes progressed to MI at a similar rate when PGE(2), PGF(2alpha) or AngII was present in the co-culture system with follicular cells (PGE(2) 77.4%, PGF(2alpha) 70.0%, and AngII 75.0% MI). In conclusion, our results provide strong evidence that AngII mediates the resumption of meiosis induced by an LH surge in bovine oocytes and that this event is dependent on PGE(2) or PGF(2alpha) produced by follicular cells.


Assuntos
Dinoprosta/metabolismo , Dinoprostona/metabolismo , Oócitos/citologia , Oócitos/metabolismo , Folículo Ovariano/metabolismo , Angiotensina II/antagonistas & inibidores , Angiotensina II/farmacologia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Bovinos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Técnicas de Cocultura , Inibidores de Ciclo-Oxigenase/farmacologia , Feminino , Hormônio Liberador de Gonadotropina/farmacologia , Indometacina/farmacologia , Hormônio Luteinizante/metabolismo , Meiose/efeitos dos fármacos , Saralasina/farmacologia
13.
Ciênc. rural ; 36(5): 1501-1506, set.-out. 2006. tab
Artigo em Inglês | LILACS | ID: lil-442521

RESUMO

The objective of this study was to determine the effects of vacuum-cooled liquid nitrogen on the development of vitrified immature (germinal vesicle stage; GV) and mature (metaphase II; MII) bovine oocytes after re-warming. Liquid nitrogen was exposed to either atmospheric pressure or to a vacuum (300mm Hg for 45sec); the latter decreased the temperature of the liquid nitrogen to -200°C. Partially denuded oocytes were vitrified either just after selection (GV) or after 22 hours of in vitro maturation (MII) in TCM 199 medium + 10 percent of estrous mare serum. For vitrification, oocytes were firstly exposed to an intermediate solution (10 percent EG + 10 percent DMSO) for 30sec, followed by the vitrification solution (20 percent EG + 20 percent DMSO + 0.5M sucrose) for 20sec. Groups of three or four oocytes were loaded into an open-pulled-straw and directly plunged into liquid nitrogen. Oocytes were subsequently re-warmed by exposure to air (25°C) for 4sec, followed by 5 min exposure to decreasing concentrations (0.3 and 0.15M) of sucrose. Fertilization (Day 0) was done with 2 x 106 spermatozoa mL-1 (selected by a swim-up procedure) and incubated for 18 to 22 hours. Presumptive zygotes were cultured at 39°C in four-well dishes with SOFaaci medium, under 5 percent CO2 and saturated humidity. Cleavage (Day 2) and blastocyst rates (Day 8) were 33.9 and 4.2 percent, respectively, for GV stage oocytes at atmospheric pressure, 41.2 and 8.8 percent for GV oocytes under vacuum, 43.5 and 6.7 percent for MII oocytes at atmospheric pressure, and 53.6 and 10.6 percent for MII oocytes under vacuum. In conclusion, vacuum-cooled liquid nitrogen improved developmental rates of vitrified-thawed bovine oocytes.


O objetivo deste estudo foi determinar o efeito do nitrogênio liquido super resfriado por vácuo no desenvolvimento, após reaquecimento, de oócitos bovinos vitrificados imaturos ou maturados. O nitrogênio líquido foi mantido em atmosfera normal ou submetido ao vácuo (300mm Hg por 45s) este último reduzindo a temperatura do nitrogênio para -200°C. Oócitos parcialmente desnudos foram vitrificados logo após a seleção (estádio de vesícula germinativa; VG), ou após 22 horas de maturação (metáfase II; MII) em meio TCM 199 + 10 por cento de soro de égua em estro. Para a vitrificação, os oócitos foram inicialmente expostos a uma solução intermediária (10 por cento EG + 10 por cento DMSO) por 30s e a seguir a uma solução de vitrificação (20 por cento EG + 20 por cento DMSO + 0,5M sacarose) por 20s. Grupos de 3 ou 4 oócitos foram envasados em palhetas estiradas e abertas e mergulhados no nitrogênio líquido. Os oócitos foram então reaquecidos por exposição ao ar (25°C) por 4s, seguido de exposição a concentrações decrescentes de sacarose (0,3 e 0,15M - 5 minutos cada). A fecundação (dia 0) foi realizada com 2 x 106 espermatozóides mL-1 (selecionados por "swim-up") e incubação por 18 a 22 horas. Os presumíveis zigotos foram cultivados a 39°C, em placas de quatro poços, com meio SOFaaci, com 5 por cento de CO2 e umidade saturada. As taxas de clivagem (Dia 2) e de blastocistos (Dia 8) obtidas foram de 33,9 e de 4,2 por cento, respectivamente, para oócitos no estágio de VG / pressão normal, de 41,2 e 8,8 por cento para oócitos VG / vácuo, 43,5 e 6,7 por cento para oócitos MII / pressão normal e de de 53,6 e 10,6 por cento para oócitos MII / vácuo. Conclui-se que o emprego de nitrogênio líquido super resfriado pelo vácuo melhora as taxas de desenvolvimento de oócitos bovinos após a vitrificação.

14.
Theriogenology ; 66(9): 2068-76, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16889824

RESUMO

The objective was to evaluate the effects of angiotensin II (Ang II), insulin-like growth factor-I (IGF-I) and insulin on the nuclear and cytoplasmic maturation of bovine oocytes in the presence of follicular cells. Cumulus-oocyte complexes (COCs) were cultured for 22h in the presence of follicular cells (control with cells) and Ang II, IGF-I or insulin (treatments), or in the absence of follicular cells (control without cells). Using these five groups, Experiment 1 was conducted with and without the addition of gonadotrophins. Only oocytes in the Ang II group resumed meiosis at rates (88.2+/-1.8% and 90.7+/-4.3% for oocytes cultured in the absence or presence of LH/FSH, respectively) similar to those observed for oocytes cultured in the absence of follicular cells (89.7+/-0.3% and 92.6+/-2.6%; P<0.01). In Experiment 2, the effect of Ang II alone and in combination with IGF-I or insulin on oocyte maturation for 7h (germinal vesicle breakdown), 12h (metaphase I) and 22h (metaphase II) was evaluated in a design similar to that of the first experiment. Ang II plus IGF-I or insulin induced the resumption of meiosis, irrespective of the presence of gonadotrophins (P<0.01). Experiment 3 used groups similar Experiment 2 to determine the rate of subsequent embryo development, using fetal calf serum (FCS) in the culture medium. The COCs were cultured in maturation medium for 1h (1+23h), 12h (12+12h) or 24h in the presence of follicular cells and the respective treatments and for the remaining period in the absence of follicular cells to complete 24h. In Experiment 4, BSA was used in lieu of serum in the maturation medium in a 12+12h maturation system. Oocytes matured using the 12+12h system with BSA or FCS in the presence of Ang II+IGF-I had higher rates of blastocyst formation than the other treatments (P<0.05). In conclusion, Ang II reversed the inhibitory effect of follicular cells on nuclear maturation of bovine oocytes, irrespective of the presence of gonadotrophins, IGF-I and insulin. However, oocyte cytoplasmatic maturation (i.e., subsequent embryo development), was higher when Ang II and IGF-I were present in the maturation medium containing follicular cells cultured for 12+12h.


Assuntos
Angiotensina II/farmacologia , Bovinos/fisiologia , Desenvolvimento Embrionário/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/farmacologia , Insulina/farmacologia , Oócitos , Animais , Bovinos/embriologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultura , Feminino , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Folículo Ovariano/citologia , Folículo Ovariano/fisiologia , Gravidez , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...