Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 827
Filtrar
1.
J Reprod Dev ; 69(4): 223-226, 2023 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-37331813

RESUMO

Superovulation procedures are routinely and widely used in mouse reproductive technology. Previous studies have shown that a large number of oocytes can be obtained from adult mice (> 10 weeks old) using a combined treatment with progesterone (P4) and anti-inhibin serum (AIS). However, these effects have not been fully investigated in young (4 weeks) C57BL/6J mice. Here, we found that a modified superovulation protocol (combined treatment with P4, AIS, eCG (equine chorionic gonadotropin), and hCG (human chorionic gonadotropin); P4D2-Ae-h) improved the number of oocytes compared to the control (eCG and hCG) (39.7 vs. 21.3 oocytes/mouse). After in vitro fertilization, pronuclear formation rates were 69.3% (P4D2-Ae-h group) and 66.2% (control group). After embryo transfer, 46.4% (116/250) of the embryos in the P4D2-Ae-h group successfully developed to term, which was comparable to the control group (42.9%; 123/287 embryos). In conclusion, our protocol (P4D2-Ae-h) was effective for superovulation in young C57BL/6J mice.


Assuntos
Gonadotropinas Equinas , Inibinas , Oócitos , Progesterona , Animais , Feminino , Humanos , Camundongos , Gonadotropina Coriônica/farmacologia , Gonadotropinas Equinas/farmacologia , Cavalos , Inibinas/farmacologia , Camundongos Endogâmicos C57BL , Progesterona/farmacologia , Superovulação
2.
Artigo em Inglês | MEDLINE | ID: mdl-36201267

RESUMO

BACKGROUND: Neuroinflammation and cytokines play critical roles in neuropathic pain and axon degeneration/regeneration. Cytokines of transforming growth factor-ß superfamily have implications in pain and injured nerve repair processing. However, the transcriptional profiles of the transforming growth factor-ß superfamily members in dorsal root ganglia under neuropathic pain and axon degeneration/regeneration conditions remain elusive. OBJECTIVE: We aimed to plot the transcriptional profiles of transforming growth factor-ß superfamily components in lumbar dorsal root ganglia of sciatic nerve-axotomized rats and to further verify the profiles by testing the analgesic effect of activin C, a representative cytokine, on neuropathic pain. METHODS: Adult male rats were axotomized in sciatic nerves, and lumbar dorsal root ganglia were isolated for total RNA extraction or section. A custom microarray was developed and employed to plot the gene expression profiles of transforming growth factor-ß superfamily components. Realtime RT-PCR was used to confirm changes in the expression of activin/inhibin family genes, and then in situ hybridization was performed to determine the cellular locations of inhibin α, activin ßC, BMP-5 and GDF-9 mRNAs. The rat spared nerve injury model was performed, and a pain test was employed to determine the effect of activin C on neuropathic pain. RESULTS: The expression of transforming growth factor-ß superfamily cytokines and their signaling, including some receptors and signaling adaptors, were robustly upregulated. Activin ßC subunit mRNAs were expressed in the small-diameter dorsal root ganglion neurons and upregulated after axotomy. Single intrathecal injection of activin C inhibited neuropathic pain in spared nerve injury model. CONCLUSION: This is the first report to investigate the transcriptional profiles of members of transforming growth factor-ß superfamily in axotomized dorsal root ganglia. The distinct cytokine profiles observed here might provide clues toward further study of the role of transforming growth factor-ß superfamily in the pathogenesis of neuropathic pain and axon degeneration/regeneration after peripheral nerve injury.


Assuntos
Neuralgia , Fator de Crescimento Transformador beta , Ratos , Masculino , Animais , Axotomia , Ratos Sprague-Dawley , Fator de Crescimento Transformador beta/farmacologia , Ativinas/genética , Ativinas/farmacologia , Nervo Isquiático/lesões , Nervo Isquiático/patologia , Neuralgia/genética , Neuralgia/patologia , RNA Mensageiro/genética , Inibinas/farmacologia , Fatores de Crescimento Transformadores/farmacologia
3.
In Vitro Cell Dev Biol Anim ; 58(3): 243-254, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35378691

RESUMO

Cytosine-phosphate-guanine oligodeoxynucleotides (CpG-ODNs), which exist in vertebrate, bacterial, and viral genomes, are regarded as strong immune adjuvants. To date, the biological activities of CpG-ODNs in reproduction remain unknown. Here, we investigated the effects of CpG-ODNs on the cell cycle, apoptosis, and steroidogenesis in mouse granulosa cells (mGCs), in combination with inhibin alpha (1 ~ 32) fragments. mGCs were transfected with pEGFP (containing green fluorescent protein, as a control), pEGISI (containing inhibin alpha (1 ~ 32) fragments), or pEGISI-CpG-ODNs (containing inhibin alpha (1 ~ 32) fragments and CpG-ODNs motifs) plasmid for 48 h in vitro. Our results showed that the mRNA and protein expression levels of inhibin alpha were downregulated in mGCs transfected with pEGISI-CpG-ODNs, compared to those transfected with pEGISI. Flow cytometry demonstrated that pEGISI-CpG-ODNs transfection promoted cell proliferation (for example, increasing the number of cells in S and G2 phases) and decreased apoptosis, compared to pEGISI transfection. The present study also indicated that the expression of cell cycle-related genes (cyclin D2, cyclin D3, cyclin E1, Cdk2, and Cdk6) was increased, while the expression of apoptosis-related factors (Fas, FasL, caspase-8, and caspase-3) decreased after pEGISI-CpG-ODNs treatment. Additionally, pEGISI-CpG-ODNs reversed the effect of pEGISI on the secretion of estradiol in mGCs, which was further validated by upregulating the levels of its synthesis-related factors (StAR, Cyp11a1, and 17ß-HSD II). Nevertheless, pEGISI-CpG-ODNs or pEGISI did not affect the concentration of progesterone nor changed the expression levels of its synthesis-related factors (3ß-HSD I and Cyp19a1). In conclusion, this study demonstrated that CpG-ODNs may affect the cell cycle, apoptosis, and steroidogenesis by targeting the effects of inhibin alpha (1 ~ 32) fragments, supporting the potential role of CpG-ODNs in the development of granulosa cells.


Assuntos
Citosina , Oligodesoxirribonucleotídeos , Animais , Apoptose/fisiologia , Ciclo Celular , Citosina/metabolismo , Citosina/farmacologia , Feminino , Células da Granulosa/metabolismo , Guanina/metabolismo , Guanina/farmacologia , Inibinas/genética , Inibinas/metabolismo , Inibinas/farmacologia , Camundongos , Oligodesoxirribonucleotídeos/metabolismo , Fosfatos/metabolismo , Fosfatos/farmacologia
4.
J Assist Reprod Genet ; 38(1): 55-69, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33067741

RESUMO

PURPOSE: Oocyte quality and reproductive outcome are negatively affected by advanced maternal age, ovarian stimulation and method of oocyte maturation during assisted reproduction; however, the mechanisms responsible for these associations are not fully understood. The aim of this study was to compare the effects of ageing, ovarian stimulation and in-vitro maturation on the relative levels of transcript abundance of genes associated with DNA repair during the transition of germinal vesicle (GV) to metaphase II (MII) stages of oocyte development. METHODS: The relative levels of transcript abundance of 90 DNA repair-associated genes was compared in GV-stage and MII-stage oocytes from unstimulated and hormone-stimulated ovaries from young (5-8-week-old) and old (42-45-week-old) C57BL6 mice. Ovarian stimulation was conducted using pregnant mare serum gonadotropin (PMSG) or anti-inhibin serum (AIS). DNA damage response was quantified by immunolabeling of the phosphorylated histone variant H2AX (γH2AX). RESULTS: The relative transcript abundance in DNA repair genes was significantly lower in MII oocytes compared to GV oocytes in young unstimulated and PMSG stimulated but was higher in AIS-stimulated mice. Interestingly, an increase in the relative level of transcript abundance of DNA repair genes was observed in MII oocytes from older mice in unstimulated, PMSG-stimulated and AIS-stimulated mice. Decreased γH2AX levels were found in both GV oocytes (82.9%) and MII oocytes (37.5%) during ageing in both ovarian stimulation types used (PMSG/AIS; p < 0.05). CONCLUSIONS: In conclusion, DNA repair relative levels of transcript abundance are altered by maternal age and the method of ovarian stimulation during the GV-MII transition in oocytes.


Assuntos
Dano ao DNA/efeitos dos fármacos , Histonas/genética , Oócitos/crescimento & desenvolvimento , Oogênese/efeitos dos fármacos , Envelhecimento/efeitos dos fármacos , Envelhecimento/genética , Envelhecimento/patologia , Animais , Reparo do DNA/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Gonadotropinas Equinas/farmacologia , Humanos , Inibinas/farmacologia , Metáfase/genética , Camundongos , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Indução da Ovulação/métodos , Gravidez
5.
Endocrinology ; 161(8)2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32569368

RESUMO

Ovarian-derived inhibin A and inhibin B (heterodimers of common α- and differing ß-subunits) are secreted throughout the menstrual cycle in a discordant pattern, with smaller follicles producing inhibin B, whereas the dominant follicle and corpus luteum produce inhibin A. The classical function for endocrine inhibins is to block signalling by activins (homodimers of ß-subunits) in gonadotrope cells of the anterior pituitary and, thereby, inhibit the synthesis of FSH. Whether inhibin A and inhibin B have additional physiological functions is unknown, primarily because producing sufficient quantities of purified inhibins, in the absence of contaminating activins, for preclinical studies has proven extremely difficult. Here, we describe novel methodology to enhance inhibin A and inhibin B activity and to produce these ligands free of contaminating activins. Using computational modeling and targeted mutagenesis, we identified a point mutation in the activin ß A-subunit, A347H, which completely disrupted activin dimerization and activity. Importantly, this ß A-subunit mutation had minimal effect on inhibin A bioactivity. Mutation of the corresponding residue in the inhibin ß B-subunit, G329E, similarly disrupted activin B synthesis/activity without affecting inhibin B production. Subsequently, we enhanced inhibin A potency by modifying the binding site for its co-receptor, betaglycan. Introducing a point mutation into the α-subunit (S344I) increased inhibin A potency ~12-fold. This study has identified a means to eliminate activin A/B interference during inhibin A/B production, and has facilitated the generation of potent inhibin A and inhibin B agonists for physiological exploration.


Assuntos
Inibinas , Engenharia de Proteínas/métodos , Feminino , Células HEK293 , Humanos , Inibinas/genética , Inibinas/isolamento & purificação , Inibinas/metabolismo , Inibinas/farmacologia , Proteínas de Membrana , Modelos Moleculares , Mutagênese/fisiologia , Ovário/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/isolamento & purificação , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacologia , Multimerização Proteica/genética , Estrutura Quaternária de Proteína/genética , Estrutura Terciária de Proteína/genética , Subunidades Proteicas/genética , Subunidades Proteicas/isolamento & purificação , Subunidades Proteicas/metabolismo , Subunidades Proteicas/farmacologia , Proteínas de Saccharomyces cerevisiae , Transfecção
6.
Cell Tissue Res ; 381(2): 337-350, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32377876

RESUMO

Inhibin A regulates follicular development, and its expression level is related to physiological activities, such as the recruitment, selection, and predominance during follicular development. Therefore, examining inhibin A and its regulatory effects on the reproductive performance of poultry is crucial. In this study, we measured the mRNA and protein abundances of INHA and INHBA in the chicken reproductive system and determined the hormone secretion and apoptosis of follicular granulosa cells (GCs) after being treated with inhibin A protein, and flow cytometry was performed to analyze GC apoptosis in INHA-specific small RNA interference (siRNA). We detected that INHA and INHBA were mainly expressed in chicken follicles. The highest INHA mRNA abundance was found in the fifth largest preovulatory follicle (F5) (P < 0.05). INHBA mRNA expression in the largest preovulatory follicle (F1) was significantly higher than those in other follicles (P < 0.05). Similar results were found for INHA and INHBA protein expression in those follicles (P < 0.05). Treatment with inhibin A protein increased the activity of GCs in a dose-dependent manner (P < 0.05), which was characterized by decreased gene expression of pro-apoptotic factors Bax and Caspase-3 (P < 0.05) and increased expression of proliferation genes Bcl-2 and PCNA (P < 0.05). Additionally, inhibin A significantly increased the secretion of progesterone and estradiol (P < 0.05). RNAi-mediated knockdown of INHA increased apoptosis in GCs via a Caspase-3-dependent mitochondrial pathway.


Assuntos
Apoptose/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Células da Granulosa , Inibinas/farmacologia , Animais , Células Cultivadas , Galinhas , Estradiol/metabolismo , Feminino , Células da Granulosa/citologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Progesterona/metabolismo
7.
Biol Reprod ; 101(2): 405-415, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31167231

RESUMO

Kisspeptin (encoded by the Kiss-1 gene) in the arcuate nucleus (ARC) of the hypothalamus governs the hypothalamic-pituitary-gonadal (HPG) axis by regulating pulsatile release of gonadotropin-releasing hormone (GnRH). Meanwhile, kisspeptin in the anteroventral periventricular nucleus (AVPV) region has been implicated in estradiol (E2)-induced GnRH surges. Kiss-1-expressing cell model mHypoA-55 exhibits characteristics of Kiss-1 neurons in the ARC region. On the other hand, Kiss-1 expressing mHypoA-50 cells originate from the AVPV region. In the mHypoA-55 ARC cells, activin significantly increased Kiss-1 gene expression. Follistatin alone reduced Kiss-1 expression within these cells. Interestingly, activin-induced Kiss-1 gene expression was completely abolished by follistatin. Inhibin A, but not inhibin B reduced Kiss-1 expression. Activin-increased Kiss-1 expression was also abolished by inhibin A. Pretreatment of the cells with follistatin or inhibin A significantly inhibited kisspeptin- or GnRH-induced Kiss-1 gene expression in mHypoA-55 cells. In contrast, in the mHypoA-50 AVPV cell model, activin, follistatin, and inhibin A did not modulate Kiss-1 gene expression. The subunits that compose activin and inhibin, as well as follistatin were expressed in both mHypoA-55 and mHypoA-50 cells. Expression of inhibin ßA and ßB subunits and follistatin was much higher in mHypoA-55 ARC cells. Furthermore, we found that expression of the inhibin α subunit and follistatin genes was modulated in the presence of E2 in mHypoA-55 ARC cells. The results of this study suggest that activin, follistatin, and inhibin A within the ARC region participate in the regulation of the HPG axis under the influence of E2.


Assuntos
Ativinas/farmacologia , Folistatina/farmacologia , Inibinas/farmacologia , Kisspeptinas/metabolismo , Animais , Linhagem Celular , Folistatina/genética , Folistatina/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Kisspeptinas/genética , Camundongos , Subunidades Proteicas
8.
Theriogenology ; 114: 136-142, 2018 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-29614400

RESUMO

Inhibins regulate folliculogenesis, gametogenesis and hormone secretion via endocrine, paracrine and autocrine manners, and play roles in encouraging or suppressing proliferation of cells. In order to investigate the effects of inhibin A on proliferation and apoptosis of porcine granulosa cells (GCs), GCs were isolated from ovarian follicles (3-6 mm), treated with inhibin A at different concentrations. The cell viability, mitochondrial membrane potential (MMP), expression of proliferation-related genes and cell cycle were detected. Inhibin α subunit (INHA) gene was silenced to detect the effect of down-regulation of inhibin on expression of proliferation-related genes in GCs. The results showed that cell viability was associated with inhibin A concentration and significantly enhanced by inhibin A at high doses (50 and 100 ng/mL, P < 0.05) compared to control group (0 ng/mL). Meanwhile, the MMP boosted after treated with 100 ng/mL inhibin A for 48 h. Expression of proliferating cell nuclear antigen (PCNA) (200 ng/mL) and CyclinB1 (100 and 200 ng/mL) was promoted while Caspase-3(100 and 200 ng/mL) and BAX (200 ng/mL) was inhibited in dose dependent manner after the cells were incubated with inhibin A for 24 h (P < 0.05) compared to control group, thereby the transition from G1 phase to S phase was promoted and the number of S phase cells was increased. After silencing the INHA gene expression, expression of Caspase-3 was enhanced and CyclinB1 was inhibited (P < 0.05) compared to Ri-negative group. All the results pointed to the conclusion that inhibin A promotes the proliferation of GCs while inhibits apoptosis.


Assuntos
Proliferação de Células/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Inibinas/farmacologia , Suínos , Animais , Caspase 3/genética , Caspase 3/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclina B1/genética , Ciclina B1/metabolismo , Relação Dose-Resposta a Droga , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Inibinas/administração & dosagem , Inibinas/genética , Inibinas/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
9.
J Immunother Cancer ; 5: 37, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28428886

RESUMO

BACKGROUND: Testicular cancer is the most common male neoplasm occurring in men between the ages of 20 and 34. Although germ-line testicular tumors respond favorably to current standard of care, testicular stromal cell (TSC) tumors derived from Sertoli cells or Leydig cells often fail to respond to chemotherapy or radiation therapy and have a 5-year overall survival significantly lower than the more common and more treatable germ line testicular tumors. METHODS: To improve outcomes for TSC cancer, we have developed a therapeutic vaccine targeting inhibin-α, a protein produced by normal Sertoli and Leydig cells of the testes and expressed in the majority of TSC tumors. RESULTS: We found that vaccination against recombinant mouse inhibin-α provides protection and therapy against transplantable I-10 mouse TSC tumors in male BALB/c mice. Similarly, we found that vaccination with the immunodominant p215-234 peptide of inhibin-α (Inα 215-234) inhibits the growth of autochthonous TSC tumors occurring in male SJL.AMH-SV40Tag transgenic mice. The tumor immunity and enhanced overall survival induced by inhibin-α vaccination may be passively transferred into naive male BALB/c recipients with either CD4+ T cells, B220+ B cells, or sera from inhibin-α primed mice. CONCLUSIONS: Considering the lack of any alternative effective treatment for chemo- and radiation-resistant TSC tumors, our results provide for the first time a rational basis for immune-mediated control of these aggressive and lethal variants of testicular cancer.


Assuntos
Imunoterapia/métodos , Inibinas/uso terapêutico , Neoplasias Embrionárias de Células Germinativas/prevenção & controle , Neoplasias Testiculares/prevenção & controle , Vacinação/métodos , Animais , Humanos , Inibinas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Embrionárias de Células Germinativas/patologia , Neoplasias Testiculares/patologia
10.
Theriogenology ; 86(5): 1341-6, 2016 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-27242176

RESUMO

Improvement of the superovulation technique will help to enhance the efficiency of embryo and animal production. Blocking inhibin using inhibin antiserum (IAS) is known to promote follicular development by increasing the level of FSH. Previously, we reported that coadministration of IAS and eCG produced more than 100 oocytes from a single female C57BL/6 mouse at 4 weeks old. The oocytes derived from the IAS + eCG (IASe) treatment were able to fertilize and develop normally into offspring. In this study, we examined the effect of IASe treatment on the numbers of ovulated oocytes in major inbred (A/J, BALB/cByJ, C3HeJ, DBA/2J, and FVB/NJ) and outbred (CD1) mice strains at 4 weeks old. We confirmed the fertilization and developmental ability of the IASe-derived oocytes. IASe treatment ovulated 1.5 to 3.2 times higher numbers of oocytes than eCG treatment alone. The fertilization rate of IASe-derived oocytes was similar to that of eCG-derived oocytes. In vitro and in vivo developmental rates of the embryos derived from IASe were similar to the rates of embryos derived from eCG. We have shown that superovulation by IASe is very effective in obtaining high numbers of ovulated oocytes from small numbers of oocyte donor in a number of mice strains. The superovulation technique will contribute to the archiving of cryopreserved embryos of genetically engineered mice using small numbers of donors and has the potential to produce more live animals for rederivation of the archived mouse lines in mouse repositories.


Assuntos
Soros Imunes/farmacologia , Imunoterapia , Inibinas/farmacologia , Superovulação/efeitos dos fármacos , Animais , Gonadotropina Coriônica , Feminino , Masculino , Camundongos , Camundongos Endogâmicos
12.
Endocrinology ; 156(8): 3047-57, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25961838

RESUMO

Gonadal-derived inhibin A and B are essential factors in mammalian reproduction, negatively regulating pituitary production of FSH. Inhibins are synthesized as heterodimers of α- and ß-subunits, each comprising an N-terminal pro- and C-terminal mature domain. After dimerization, the inhibin α- and ß-subunit prodomains are enzymatically cleaved from the mature domains at consensus RXXR sites (site1). Interestingly, the inhibin α-subunit is a unique TGF-ß ligand, comprising a second cleavage site (site2) within its prodomain. Cleavage at site2 in the inhibin α-subunit prodomain releases a 43-amino acid proα-peptide. We aimed to determine the influence of the proα-peptide on inhibin synthesis and bioactivity. Blocking proα-peptide release by silencing cleavage site2 (Arg56-Arg61) inhibited both inhibin A and B synthesis. Ligand blot analysis and solid-phase binding assays indicated that the proα-peptide binds specifically to a mature 30-kDa inhibin (mean Kd 86 nM) but was unable to bind related activins. The proα-peptide suppressed inhibin A and B bioactivity in primary rat pituitary cell cultures. Mechanistically, the proα-peptide blocked inhibin A binding to its coreceptor, betaglycan (IC50 131 nM), and the subsequent sequestration of the activin type II receptor (IC50 156 nM), which underscores inhibin's biological activity. Based on the sequential mutations across the inhibin α-subunit, the proα-peptide binding site was localized to residues Arg341-Thr354, corresponding directly to the betaglycan binding region. Together our findings indicate that the proα-peptide limits the synthesis and bioactivity of inhibins.


Assuntos
Inibinas/biossíntese , Inibinas/farmacologia , Fragmentos de Peptídeos/farmacologia , Precursores de Proteínas/química , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Células HEK293 , Humanos , Inibinas/antagonistas & inibidores , Inibinas/genética , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/farmacologia , Precursores de Proteínas/genética , Precursores de Proteínas/farmacologia , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Proteoglicanas/metabolismo , Ratos , Receptores de Fatores de Crescimento Transformadores beta/metabolismo
13.
Biol Reprod ; 88(6): 165, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23658431

RESUMO

Mechanisms regulating gonadotropin surges and gonadotropin requirements for follicle emergence and selection were studied in heifers. Experiment 1 evaluated whether follicular inhibins regulate the preovulatory luteinizing hormone (LH)/follicle-stimulating hormone (FSH) surges elicited by gonadotropin-releasing hormone (GnRH) injection (Hour = 0) and the subsequent periovulatory FSH surge. Treatments included control (n = 6), steroid-depleted bovine follicular fluid (bFF) at Hour -4 (n = 6), and bFF at Hour 6 (n = 6). Gonadotropins in blood were assessed hourly from Hours -6 to 36, and follicle growth tracked by ultrasound. Consistent with inhibin independence, bFF at Hour -4 did not impact the GnRH-induced preovulatory FSH surge, whereas treatment at Hour 6 delayed onset of the periovulatory FSH surge and impeded growth of a new follicular wave. Experiment 2 examined GnRH and estradiol (E2) regulation of the periovulatory FSH surge. Treatment groups were control (n = 8), GnRH-receptor antagonist (GnRHr-ant, n = 8), and E2 + GnRHr-ant (n = 4). GnRHr-ant (acyline) did not reduce the concentrations of FSH during the periovulatory surge and early follicle development (<7.0 mm) was unaffected, although subsequent growth of a dominant follicle (>8.0 mm) was prevented by GnRHr-ant. Addition of E2 delayed both the onset of the periovulatory FSH surge and emergence of a follicular wave. Failure to select a dominant follicle in the GnRHr-ant group was associated with reduced concentrations of LH but not FSH. Maximum diameter of F1 in controls (13.3 ± 0.5 mm) was greater than in both GnRHr-ant (7.7 ± 0.3 mm) and E2 + GnRHr-ant (6.7 ± 0.8 mm) groups. Results indicated that the periovulatory FSH surge stems from removal of negative stimuli (follicular E2 and inhibin), but is independent of GnRH stimulation. Emergence and early growth of follicles (until about 8 mm) requires the periovulatory FSH surge but not LH pulses. However, follicular deviation and late-stage growth of a single dominant follicle requires GnRH-dependent LH pulses.


Assuntos
Estradiol/farmacologia , Hormônio Foliculoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Inibinas/farmacologia , Hormônio Luteinizante/metabolismo , Folículo Ovariano/efeitos dos fármacos , Ovulação/fisiologia , Animais , Bovinos , Feminino , Hormônio Foliculoestimulante/sangue , Líquido Folicular/efeitos dos fármacos , Líquido Folicular/fisiologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Hormônio Luteinizante/sangue , Folículo Ovariano/diagnóstico por imagem , Folículo Ovariano/crescimento & desenvolvimento , Ovulação/efeitos dos fármacos , Ultrassonografia
14.
Res Vet Sci ; 95(2): 374-80, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23602073

RESUMO

The objective of the present study was to isolate and purify high MW inhibin (≈ 129 kDa) from buffalo ovarian follicular fluid (buFF) and to investigate its biological activity. Throughout the process of purification, the inhibin fractions were evaluated for bioactivity by a specific, sensitive and uniformly reproducible bioassay in mice. The final biological activity of this preparation was tested in normal cycling adult female Barbari goats. Eight animals, randomly divided into two groups, were synchronized for estrus by administering PGF2α twice at an interval of ten days. Following synchronization, the treatment group (n=4) received (i.m.) 0.4 ml (240 µg protein total dose) of purified inhibin (MW ≈ 129 kDa) of buFF in the morning at 08.00 h for the four consecutive days of follicular phase, while the control group (n=4) received only saline (0.4 ml). Blood samples were collected from jugular vein immediately before the first injection and subsequent collections were made daily in the afternoon until day 8 of the experiment including four days (0, 1, 2, & 3 days) of the next cycle. FSH was assayed in all the samples by ELISA. The peripheral FSH concentration sharply declined from 1.854±0.137 to 0.979 ± 0.02 u/l, 8h after the administration of inhibin on the first day. The value in controls was 2.004 ± 0.132 u/l. For the duration of treatment of four consecutive days, the FSH level in experimental group remained significantly low (p<0.05) compared to control group. After cessation of treatment, the FSH level remained low on day 0 and 1 of the next cycle in the experimental and control animals. However, a significant rebound increase in plasma FSH levels occurred on day 2 & 3 (2.73 ± 0.179 & 1.849 ± 0.128 u/l) only in the experimental group compared to control animals (P<0.05). This increment might be caused by the rebound surge of FSH from anterior pituitary which further corroborates the effect of inhibin in treated animals. In conclusion, the present study demonstrates that the high MW form of inhibin (≈ 129 kDa) isolated from buFF has comparable biological activity as revealed by 31-32 kDa inhibin from other species.


Assuntos
Búfalos/fisiologia , Hormônio Foliculoestimulante/sangue , Líquido Folicular/química , Cabras/sangue , Inibinas/farmacologia , Animais , Ciclo Estral , Feminino , Líquido Folicular/metabolismo , Cabras/metabolismo , Inibinas/química , Inibinas/metabolismo , Camundongos
15.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 43(3): 348-51, 2012 May.
Artigo em Chinês | MEDLINE | ID: mdl-22812235

RESUMO

OBJECTIVE: To study the effects of activin A (ACTA) and inhibin A (INHA) on the in vitro maturation (IVM) of mice immature eggs and early embryonic development. METHODS: 1. The mice oocytes were cultured in the culture medium contained different concentration of ACTA and INHA (50 ng/mL, 100 ng/mL, 200 ng/mL) to find the optimum concentration; 2. The effects of different concentration of ACTA, INHA, ACTA+ INHA on the mature rates of mice oocytes were compared; 3. The effects of different concentration of ACTA, INHA, ACTA+INHA on the fertility rates and blastocysts formation rates of IVM mice eggs were also compared. RESULTS: The mature rates of mice oocytes in culture medium contained (100 ng/mL and 200 ng/mL) ACTA and INHA were significantly higher than that contained 50 ng/mL ACTA and INHA (P < 0.05); There was no difference between 100 ng/mL and 200 ng/mL groups. The mature rates, fertility rates and blastocyst rates of mice oocytes were significantly higher than those of control (P < 0.05). CONCLUSION: The mature rates, fertility rates and embryonic development potential after fertilization of mice oocytes were significantly promoted by ACTA, INHA. The optimum concentration of ACTA and INHA in culture medium was 100 ng/mL.


Assuntos
Ativinas/farmacologia , Técnicas de Maturação in Vitro de Oócitos/métodos , Inibinas/farmacologia , Oócitos/efeitos dos fármacos , Animais , Células Cultivadas , Feminino , Fertilização in vitro , Masculino , Camundongos , Oócitos/citologia
16.
Anim Biotechnol ; 23(2): 71-88, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22537057

RESUMO

Inhibin is a non-steroidal glycoprotein hormone of gonadal origin with major action as negative feedback control of the production of FSH by the anterior pituitary gland. The physiological role of inhibin has led to the development of inhibin immunogens for fertility enhancement in farm animals. It is envisaged that a reduction of endogenous inhibin secretion would increase FSH concentrations and thus offers a potential for increasing the number of ovulatory follicles in the ovary. The present work was carried out to produce recombinant bovine (Indian Sahiwal Cattle; Bos indicus) alpha inhibin (bINH-α) in E. coli by optimizing its expression and purification in biologically active form and to study its immunological characterization. A bacterial protein expression vector system based on the phage T(5) promoter was used. The bINH-α encoding gene was successfully cloned and expressed in E. coli and the purified recombinant bINH-α was characterized. Recombinant bINH-α (25 µg mL(-1)) immunized guinea pigs had a significant increase in litter size compared to the control group. These results indicate a role for recombinant bINH-α as a fecundity vaccine to enhance the ovulation rate and litter size in animals.


Assuntos
Bovinos/genética , Inibinas/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Western Blotting , Bovinos/metabolismo , Clonagem Molecular , Feminino , Fertilidade/efeitos dos fármacos , Cobaias , Inibinas/química , Inibinas/genética , Inibinas/farmacologia , Tamanho da Ninhada de Vivíparos/efeitos dos fármacos , Masculino , Dados de Sequência Molecular , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Estatísticas não Paramétricas
17.
J Mol Endocrinol ; 48(1): 49-60, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22082494

RESUMO

Activin and inhibin are important local modulators of theca cell steroidogenesis in the ovary. Using a serum-free primary theca cell culture system, this study investigated the effects of inhibin on theca cell androgen production and expression of steroidogenic enzymes. Androstenedione secretion from theca cells cultured in media containing activin, inhibin and follistatin was assessed by RIA over 144 h. Activin (1-100 ng/ml) suppressed androstenedione production. Inhibin (1-100 ng/ml) blocked the suppressive effects of added activin, but increased androstenedione production when added alone, suggesting it was blocking endogenous activin produced by theca cells. Addition of SB-431542 (activin receptor inhibitor) and follistatin (500 ng/ml) increased androstenedione production, supporting this concept. Infection of theca cells with adenoviruses expressing inhibitory Smad6 or 7 increased androstenedione secretion, confirming that the suppressive effects of activin required activation of the Smad2/3 pathway. Activin decreased the expression levels of steroidogenic acute regulatory protein (STAR), whereas STAR expression was increased by inhibin and SB-431542, alone and in combination. CYP11A was unaffected. The expression of CYP17 encoding 17α-hydroxylase was unaffected by activin but increased by inhibin and SB-431542, and when added in combination the effect was further enhanced. The expression of 3ß-hydroxysteroid dehydrogenase (3ß-HSD) was significantly decreased by activin, while inhibin alone and in combination with SB-431542 both potently increased the expression of 3ß-HSD. In conclusion, activin suppressed theca cell androstenedione production by decreasing the expression of STAR and 3ß-HSD. Inhibin and other blockers of activin action reversed this effect, supporting the concept that endogenous thecal activin modulates androgen production in theca cells.


Assuntos
Ativinas/farmacologia , Androgênios/biossíntese , Inibinas/farmacologia , Esteroide Hidroxilases/metabolismo , Células Tecais/metabolismo , Receptores de Ativinas/antagonistas & inibidores , Animais , Benzamidas/farmacologia , Dioxóis/farmacologia , Feminino , Folistatina/farmacologia , Expressão Gênica/efeitos dos fármacos , Proteínas Inibidoras de Diferenciação/genética , Ovinos , Proteína Smad6/antagonistas & inibidores , Proteína Smad7/antagonistas & inibidores , Esteroide Hidroxilases/genética , Células Tecais/efeitos dos fármacos , Células Tecais/enzimologia
18.
Femina ; 39(7): 351-356, jul. 2011.
Artigo em Português | LILACS | ID: lil-613323

RESUMO

A endometriose é uma condição ginecológica, que atinge mulheres em idade reprodutiva e pode ser causa de dor e infertilidade. A patogênese da doença é multifatorial e envolve a perda da capacidade de diferenciação das células endometrióticas, moléculas de adesão celular para adesão do endométrio ao peritônio, neoangiogênese, características do fluido peritoneal e alterações do sistema imune. A superfamília do fator transformador de crescimento β (TGF-β) parece exercer papéis importantes na implantação e manutenção do tecido ectópico na endometriose. Ativinas, inibinas, folistatina, hormônio anti-mülleriano e as proteínas morfogenéticas ósseas são membros da superfamília do TGF-β. Estas moléculas são expressas no endométrio humano e apresentam ações importantes na proliferação celular, diferenciação celular, função imune, regulação da apoptose e remodelamento dos tecidos, apresentando, por conseguinte, um importante papel no ciclo menstrual, decidualização do endométrio e no início da gestação. Este artigo objetiva rever os achados sobre tais proteínas no endométrio e seus possíveis papéis na gênese e fisiopatologia da endometriose


Endometriosis is a gynecological pathological entity typical of women in reproductive age, associated with pelvic pain and infertility. The pathogenesis of the disease is multifactorial and it involves loss of the endometriotic cell differentiation, cell adhesion, neo-angiogenesis, peritoneal fluid characteristics, and changes in the immune system. The transforming growth factor β (TGF-β) superfamily seems to play important roles in the implementation and maintenance of ectopic tissue in endometriosis. Activin, inhibin, follistatin, anti-Mullerian hormone, and bone morphogenetic proteins are members of the superfamily of TGF-β. The TGF-β and family members are expressed by human endometrium and act on cell proliferation, differentiation, immune function, apoptosis and tissue remodeling, playing a role in menstrual cycle, decidualization, and early pregnancy. The aim of this study is to review the findings about these molecules in the endometrium and their possible roles in the genesis and pathophysiology of endometriosis


Assuntos
Humanos , Feminino , Ativinas/farmacologia , Ativinas/genética , Endométrio/metabolismo , Endometriose/fisiopatologia , Endometriose/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Inibinas/farmacologia , Inibinas/genética , Diferenciação Celular , Ciclo Menstrual/metabolismo , Infertilidade Feminina/etiologia , Proliferação de Células
19.
Ann N Y Acad Sci ; 1192: 153-60, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20392231

RESUMO

Accumulating evidence demonstrates increasing bone turnover and bone loss in women prior to menopause and decreases in serum estradiol levels. Increased follicle-stimulating hormone levels have been correlated with some of these peri-menopausal changes. However, decreases in gonadal inhibins of the transforming growth factor (TGF)-beta superfamily strongly correlate with increases in bone formation and resorption markers across the menopause transition and predict lumbar bone mass in peri-menopausal women, likely as a result of direct inhibin suppression of osteoblastogenesis and osteoclastogenesis. Inhibins bind specifically to cells during osteoblastogenesis and osteoclastogenesis. They can block bone morphogenetic protein (BMP)-stimulated osteoblast and osteoclast development as well as BMP-stimulated SMAD1 phosphorylation, likely via inhibin-beta-glycan sequestration of BMP Type II receptor (BMPRII). Interestingly, continuous in vivo exposure to inhibin A is anabolic and protective against gonadectomy-induced bone loss in mice, suggesting that inhibins contribute to the endocrine regulation of bone metabolism via a bimodal mechanism of action whereby cycling inhibin exposure suppresses bone turnover and continuous exposure to inhibins is anabolic.


Assuntos
Remodelação Óssea/fisiologia , Menopausa/fisiologia , Animais , Biomarcadores/sangue , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Feminino , Humanos , Inibinas/farmacologia , Inibinas/fisiologia , Menopausa/sangue , Menopausa/metabolismo , Camundongos , Modelos Biológicos , Osteoblastos/efeitos dos fármacos , Osteoblastos/fisiologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/fisiologia
20.
Mol Endocrinol ; 24(3): 608-20, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20160125

RESUMO

Inhibin is an atypical member of the TGFbeta family of signaling ligands and is classically understood to function via competitive antagonism of activin ligand binding. Inhibin-null (Inha-/-) mice develop both gonadal and adrenocortical tumors, the latter of which depend upon gonadectomy for initiation. We have previously shown that gonadectomy initiates adrenal tumorigenesis in Inha-/- mice by elevating production of LH, which drives aberrant proliferation and differentiation of subcapsular adrenocortical progenitor cells. In this study, we demonstrate that LH signaling specifically up-regulates expression of TGFbeta2 in the subcapsular region of the adrenal cortex, which coincides with regions of aberrant Smad3 activation in Inha-/- adrenal glands. Consistent with a functional interaction between inhibin and TGFbeta2, we further demonstrate that recombinant inhibin-A antagonizes signaling by TGFbeta2 in cultured adrenocortical cells. The mechanism of this antagonism depends upon the mutual affinity of inhibin-A and TGFbeta2 for the signaling coreceptor betaglycan. Although inhibin-A cannot physically displace TGFbeta2 from its binding sites on betaglycan, binding of inhibin-A to the cell surface causes endocytic internalization of betaglycan, thereby reducing the number of available binding sites for TGFbeta2 on the cell surface. The mechanism by which inhibin-A induces betaglycan internalization is clathrin independent, making it distinct from the mechanism by which TGFbeta ligands themselves induce betaglycan internalization. These data indicate that inhibin can specifically antagonize TGFbeta2 signaling in cellular contexts where surface expression of betaglycan is limiting and provide a novel mechanism for activin-independent phenotypes in Inha-/- mice.


Assuntos
Inibinas/farmacologia , Inibinas/fisiologia , Proteoglicanas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Córtex Suprarrenal/metabolismo , Animais , Células Cultivadas , Citometria de Fluxo , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Humanos , Immunoblotting , Hibridização In Situ , Inibinas/genética , Inibinas/metabolismo , Camundongos , Camundongos Mutantes , Proteoglicanas/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...