Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Assist Reprod Genet ; 40(3): 443-454, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36542312

RESUMO

PURPOSE: As a secondary report to elucidate the diverse spectrum of oncofertility practices for childhood cancer around the globe, we present and discuss the comparisons of oncofertility practices for childhood cancer in limited versus optimum resource settings based on data collected in the Repro-Can-OPEN Study Part I & II. METHODS: We surveyed 39 oncofertility centers including 14 in limited resource settings from Africa, Asia, and Latin America (Repro-Can-OPEN Study Part I), and 25 in optimum resource settings from the USA, Europe, Australia, and Japan (Repro-Can-OPEN Study Part II). Survey questions covered the availability of fertility preservation and restoration options offered in case of childhood cancer as well as their degree of utilization. RESULTS: In the Repro-Can-OPEN Study Part I & II, responses for childhood cancer and calculated oncofertility scores showed the following characteristics: (1) higher oncofertility scores in optimum resource settings than in limited resource settings for ovarian and testicular tissue cryopreservation; (2) frequent utilization of gonadal shielding, fractionation of anticancer therapy, oophoropexy, and GnRH analogs; (3) promising utilization of oocyte in vitro maturation (IVM); and (4) rare utilization of neoadjuvant cytoprotective pharmacotherapy, artificial ovary, in vitro spermatogenesis, and stem cells reproductive technology as they are still in preclinical or early clinical research settings. CONCLUSIONS: Based on Repro-Can-OPEN Study Part I & II, we presented a plausible oncofertility best practice model to help optimize care for children with cancer in various resource settings. Special ethical concerns should be considered when offering advanced and innovative oncofertility options to children.


Assuntos
Preservação da Fertilidade , Neoplasias , Masculino , Feminino , Humanos , Preservação da Fertilidade/métodos , Criopreservação , Neoplasias/complicações , Neoplasias/terapia , Inquéritos e Questionários , Austrália
2.
J Assist Reprod Genet ; 39(8): 1693-1712, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35870095

RESUMO

Since 2007, the Oncofertility Consortium Annual Conference has brought together a diverse network of individuals from a wide range of backgrounds and professional levels to disseminate emerging basic and clinical research findings in fertility preservation. This network also developed enduring educational materials to accelerate the pace and quality of field-wide scientific communication. Between 2007 and 2019, the Oncofertility Consortium Annual Conference was held as an in-person event in Chicago, IL. The conference attracted approximately 250 attendees each year representing 20 countries around the world. In 2020, however, the COVID-19 pandemic disrupted this paradigm and precluded an in-person meeting. Nevertheless, there remained an undeniable demand for the oncofertility community to convene. To maintain the momentum of the field, the Oncofertility Consortium hosted a day-long virtual meeting on March 5, 2021, with the theme of "Oncofertility Around the Globe" to highlight the diversity of clinical care and translational research that is ongoing around the world in this discipline. This virtual meeting was hosted using the vFairs ® conference platform and allowed over 700 people to participate, many of whom were first-time conference attendees. The agenda featured concurrent sessions from presenters in six continents which provided attendees a complete overview of the field and furthered our mission to create a global community of oncofertility practice. This paper provides a synopsis of talks delivered at this event and highlights the new advances and frontiers in the fields of oncofertility and fertility preservation around the globe from clinical practice and patient-centered efforts to translational research.


Assuntos
COVID-19 , Preservação da Fertilidade , Neoplasias , COVID-19/epidemiologia , Humanos , Pandemias
3.
Ann Oncol ; 30(11): 1760-1775, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31418765

RESUMO

Oncofertility is a new interdisciplinary field at the intersection of oncology and reproductive medicine that expands fertility options for young cancer patients. The most common forms of hematological malignancies that occur in girls and young women and therefore necessitate oncofertility care are acute lymphocytic leukemia, acute myeloid leukemia, non-Hodgkin's lymphoma, and Hodgkin's lymphoma. Aggressive gonadotoxic anticancer regimens including alkylating chemotherapy and total body irradiation are used often in treating girls and young women with hematological malignancies. The risks of gonadotoxicity and subsequent iatrogenic premature ovarian insufficiency and fertility loss depend mainly on the type and stage of the disease, dose of anticancer therapy as well as the age of the patient at the beginning of treatment. To avoid or at least mitigate the devastating complications of anticancer therapy-induced gonadotoxicity, effective and comprehensive strategies that integrate different options for preserving and restoring fertility ranging from established to experimental strategies should be offered before, during, and after chemotherapy or radiotherapy. A multidisciplinary approach that involves strong coordination and collaboration between hemato-oncologists, gynecologists, reproductive biologists, research scientists, and patient navigators is essential to guarantee high standard of care.


Assuntos
Preservação da Fertilidade/métodos , Neoplasias Hematológicas/terapia , Oncologia/métodos , Insuficiência Ovariana Primária/etiologia , Medicina Reprodutiva/métodos , Adolescente , Adulto , Fatores Etários , Antineoplásicos Alquilantes/efeitos adversos , Sobreviventes de Câncer , Criança , Pré-Escolar , Feminino , Fertilidade/efeitos dos fármacos , Fertilidade/efeitos da radiação , Preservação da Fertilidade/normas , Neoplasias Hematológicas/mortalidade , Humanos , Lactente , Recém-Nascido , Colaboração Intersetorial , Oncologia/organização & administração , Oncologia/normas , Equipe de Assistência ao Paciente/organização & administração , Equipe de Assistência ao Paciente/normas , Medicina Reprodutiva/organização & administração , Medicina Reprodutiva/normas , Padrão de Cuidado , Taxa de Sobrevida , Resultado do Tratamento , Irradiação Corporal Total/efeitos adversos , Adulto Jovem
5.
Zygote ; 23(4): 475-84, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24666604

RESUMO

The aim of this study was to evaluate the influence of two-dimensional (2D) and three-dimensional (3D) alginate culture systems on in vitro development of pre-antral caprine follicles. In addition, the influence of the reproductive age of the ovary donor on the in vitro culture success was investigated. Pre-antral follicles from pre-pubertal or adult goats were isolated and cultured directly on a plastic surface (2D) or encapsulated in an alginate-based matrix (3D). After 18 days, the oocytes underwent in vitro maturation (IVM) and in vitro fertilization (IVF) to produce embryos. The 3D system showed higher rates of follicle survival, lower rates of oocyte extrusion, and a greater number of recovered oocytes for IVM and IVF (P < 0.05). Only pre-antral follicles from adult animals produced MII oocytes and embryos. The estradiol concentrations increased from day 2 to day 12 of culture in all groups tested (P < 0.05). Conversely, progesterone concentrations were lower in 3D-cultured follicles than in 2D-cultured follicles, with differences on days 2 and 6 of culture (P < 0.05). We provide compelling evidence that a 2D or 3D alginate in vitro culture system offers a promising approach to achieving full in vitro development of caprine pre-antral follicles to produce mature oocytes that are capable of fertilization and viable embryos.


Assuntos
Técnicas de Cultura de Células/métodos , Oócitos/fisiologia , Folículo Ovariano/crescimento & desenvolvimento , Fatores Etários , Alginatos , Animais , Técnicas de Cultura de Células/instrumentação , Sobrevivência Celular , Estradiol/metabolismo , Feminino , Fertilização in vitro , Ácido Glucurônico , Cabras , Ácidos Hexurônicos , Técnicas de Maturação in Vitro de Oócitos/métodos , Masculino , Oócitos/citologia , Folículo Ovariano/fisiologia , Puberdade
6.
Mol Hum Reprod ; 20(11): 1077-89, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25143461

RESUMO

Rapid cellular zinc influx regulates early mammalian development during the oocyte-to-egg transition through modulation of the meiotic cell cycle. Despite the physiological necessity of this zinc influx, the molecular mechanisms that govern such accumulation are unknown. Here we show that the fully grown mammalian oocyte does not employ a transcriptionally based mechanism of zinc regulation involving metal response element-binding transcription factor-1 (MTF-1), as demonstrated by a lack of MTF-1 responsiveness to environmental zinc manipulation. Instead, the mammalian oocyte controls zinc uptake through two maternally derived and cortically distributed zinc transporters, ZIP6 and ZIP10. Targeted disruption of these transporters using several approaches during meiotic maturation perturbs the intracellular zinc quota and results in a cell cycle arrest at a telophase I-like state. This arrest phenocopies established models of zinc insufficiency during the oocyte-to-egg transition, indicating the essential function of these maternally expressed transporters. Labile zinc localizes to punctate cytoplasmic structures in the human oocyte, and ZIP6 and ZIP10 are enriched in the cortex. Altogether, we demonstrate a mechanism of metal regulation required for female gamete development that may be evolutionarily conserved.


Assuntos
Proteínas de Transporte de Cátions/fisiologia , Zinco/metabolismo , Adolescente , Adulto , Animais , Transporte Biológico/genética , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Ciclo Celular/genética , Feminino , Regulação da Expressão Gênica , Homeostase , Humanos , Camundongos Endogâmicos , Oócitos/metabolismo , Elementos de Resposta , Telófase
7.
Mol Hum Reprod ; 19(12): 828-37, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24026057

RESUMO

In women, ovary and adrenal gland produce androgens. Androgens are essential drivers of the primordial to antral follicle development, prior to serving as substrate for estrogen production in the later stages of folliculogenesis. Androgens play a crucial role in the follicular-stromal intertalk by fine tuning the extracellular matrix and vessel content of the ovarian stroma. Local auto-and paracrine factors regulate androgen synthesis in the pre-antral follicle. Androgen excess is a hallmark of polycystic ovary syndrome and is a key contributor in the exaggerated antral follicle formation, stromal hyperplasia and hypervascularity. Hyperandrogenaemia overrides the follicular-stromal dialog, resulting in follicular arrest and disturbed ovulation. On the other hand, androgen deficiency is likely to have a negative impact on fertility as well, and further research is needed to examine the benefits of androgen-replacement therapy in subfertility.


Assuntos
Androgênios/fisiologia , Folículo Ovariano/crescimento & desenvolvimento , Síndrome do Ovário Policístico/etiologia , Feminino , Humanos , Infertilidade Feminina/complicações , Infertilidade Feminina/metabolismo , Doenças Ovarianas/complicações , Doenças Ovarianas/metabolismo , Doenças Ovarianas/patologia , Folículo Ovariano/metabolismo , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/patologia , Receptores Androgênicos/metabolismo
8.
Reproduction ; 145(1): 19-32, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23108112

RESUMO

In vitro follicle growth in alginate hydrogels is a unique and versatile method for studying ovarian and follicle biology that may also have implications for fertility preservation. Current culture systems support the development of isolated mouse follicles from the secondary stage onward. However, it has been a challenge to grow smaller follicles in vitro due to the dissociation of the oocyte from companion somatic cells. Recent work has demonstrated that coculturing primary follicles with mouse embryonic fibroblasts or ovarian stromal cells supports follicle survival and growth. In this study, we demonstrate that follicles themselves can exert a beneficial coculture effect. When primary follicles were cultured in groups of five or ten (multiple follicle culture), there was increased growth and survival. The multiple follicle culture approach maintained follicle integrity and resulted in the formation of antral stage follicles containing meiotically competent gametes. The growth and survival of primary follicles were highly number dependent, with the most significant enhancement observed when the largest number of follicles was grown together. Our data suggest that the follicle unit is necessary to produce the secreted factors responsible for the supportive effects of multiple follicle culture, as neither denuded oocytes, oocyte-secreted factors, nor granulosa cells alone were sufficient to support early follicle growth in vitro. Therefore, there may be signaling from both the oocyte and the follicle that enhances growth but requires both components in a feedback mechanism. This work is consistent with current in vivo models for follicle growth and thus advances the movement to recapitulate the ovarian environment in vitro.


Assuntos
Técnicas de Cocultura , Folículo Ovariano/citologia , Comunicação Parácrina/fisiologia , Animais , Comunicação Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Feminino , Camundongos , Camundongos Endogâmicos , Modelos Animais , Oócitos/citologia , Oócitos/fisiologia , Folículo Ovariano/fisiologia
9.
Nat Genet ; 25(4): 453-7, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10932194

RESUMO

The activins (dimers of betaA or betaB subunits, encoded by the genes Inhba and Inhbb, respectively) are TGF-beta superfamily members that have roles in reproduction and development. Whereas mice homozygous for the Inhba-null allele demonstrate disruption of whisker, palate and tooth development, leading to neonatal lethality, homozygous Inhbb-null mice are viable, fertile and have eye defects. To determine if these phenotypes were due to spatiotemporal expression differences of the ligands or disruption of specific ligand-receptor interactions, we replaced the region of Inhba encoding the mature protein with Inhbb, creating the allele Inhbatm2Zuk (hereafter designated InhbaBK). Although the craniofacial phenotypes of the Inhba-null mutation were rescued by the InhbaBK allele, somatic, testicular, genital and hair growth were grossly affected and influenced by the dosage and bioactivity of the allele. Thus, functional compensation within the TGF-beta superfamily can occur if the replacement gene is expressed appropriately. The novel phenotypes in these mice further illustrate the usefulness of insertion strategies for defining protein function.


Assuntos
Ativinas , Subunidades beta de Inibinas , Inibinas/genética , Oligopeptídeos , Peptídeos/genética , Alelos , Animais , Peso Corporal , Células CHO , Células Cultivadas , Cricetinae , DNA Recombinante , Embrião de Mamíferos/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Hibridização In Situ , Inibinas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Mutantes , Mutagênese Insercional , Mutação , Tamanho do Órgão , Ovário/metabolismo , Fenótipo , Gravidez , RNA/genética , RNA/metabolismo , Análise de Sobrevida , Testículo/crescimento & desenvolvimento , Testículo/metabolismo
10.
Hum Reprod ; 27(6): 1801-10, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22456922

RESUMO

BACKGROUND: In vitro follicle growth is a promising fertility preservation strategy in which ovarian follicles are cultured to produce mature and fertilization-competent oocytes. However, in primates, there has been limited success with in vitro follicle growth starting from primordial and primary follicles because adequate isolation methods and culture strategies have not been established. Understanding how to use primordial follicles for fertility preservation has significant implications because these follicles are the most abundant in the ovary, are found in all females and are fairly resistant to cryopreservation and chemotherapeutics. METHODS: In the primate ovary, primordial follicles are concentrated near the collagen-rich ovarian cortex. To obtain these follicles, we separated the ovarian cortex prior to enzymatic digestion and enriched the primordial follicle concentration by using a novel double filtration system. To test the hypothesis that a rigid physical environment, as found in vivo, is optimal for survival, primordial follicles were cultured in different concentrations of alginate for up to 6 days. Follicle survival and morphology were monitored throughout the culture. RESULTS: We found that primate ovarian tissue can be maintained for up to 24 h at 4°C without compromising tissue or follicle health. Hundreds of intact and viable primordial follicles were isolated from each ovary independent of animal age. Follicle survival and morphology were more optimal when follicles were cultured in 2% alginate compared with 0.5% alginate. CONCLUSIONS: By mimicking the rigid ovarian environment through the use of biomaterials, we have established conditions that support primordial follicle culture. These results lay the foundations for studying the basic biology of primordial follicles in a controlled environment and for using primordial follicles for fertility preservation methods.


Assuntos
Macaca mulatta , Folículo Ovariano/fisiologia , Técnicas de Cultura de Tecidos/veterinária , Alginatos , Animais , Meios de Cultura , Feminino , Ácido Glucurônico , Ácidos Hexurônicos , Folículo Ovariano/crescimento & desenvolvimento , Ovário/fisiologia , Técnicas de Cultura de Tecidos/métodos , Coleta de Tecidos e Órgãos/métodos , Coleta de Tecidos e Órgãos/veterinária
11.
Hum Reprod ; 26(9): 2473-85, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21669966

RESUMO

BACKGROUND: Ovarian tissue cryopreservation is an emerging fertility preservation option, and culturing follicles isolated from this tissue to obtain mature gametes may ultimately be the best solution for patients for whom transplantation is contraindicated. It is unclear, however, how patient-specific variables (including age, weight and menstrual cycle stage) impact follicle growth and quality during three-dimensional culture. METHODS: We used a mouse model to systematically determine how these variables impact in vitro follicle growth. We characterized metabolic and hormonal profiles of mice at specific ages, weights and cycle stages and secondary follicles from these cohorts were isolated and cultured. We then assessed follicle survival, growth and function, as well as meiotic competence and spindle morphology of the resulting oocytes. RESULTS: We found that older mice and mice with increased body weight had higher serum cholesterol, abnormal glucose tolerance and lower levels of circulating Anti-Müllerian hormone compared with younger and leaner controls. Secondary follicles isolated from different cohorts and grown in vitro had indistinguishable growth trajectories. However, the follicles isolated from older and heavier mice and those in diestrus had altered hormone profiles. These follicles contained oocytes with reduced meiotic competence and produced oocytes with greater spindle defects. CONCLUSIONS: These results suggest that the original physical environment of the follicle within the ovary can impact its function when isolated and cultured. These findings are valuable as we begin to use in vitro follicle growth technology for a heterogeneous fertility preservation patient population.


Assuntos
Ciclo Estral , Folículo Ovariano/fisiologia , Técnicas de Cultura de Tecidos , Fatores Etários , Animais , Hormônio Antimülleriano/sangue , Peso Corporal , Colesterol/sangue , Feminino , Preservação da Fertilidade , Glucose/metabolismo , Teste de Tolerância a Glucose , Meiose/fisiologia , Camundongos , Camundongos Endogâmicos , Oócitos/citologia , Oócitos/fisiologia , Oócitos/ultraestrutura , Fuso Acromático/ultraestrutura
12.
Reproduction ; 142(1): 113-22, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21502334

RESUMO

The present study examined the influences of the physical and hormonal microenvironment on in vitro growth and steroidogenesis of dog follicles. Follicles were enzymatically isolated and individually encapsulated in 0.5% (w/v; n=17) or 1.5% (n=10) alginate and cultured with 0.5 IU/ml equine chorionic gonadotropin for 192 h. In a separate experiment, follicles were encapsulated in 0.5% alginate and cultured with 0 (n=22), 1 (n=23), 10 (n=20) or 100 (n=21) µg/ml FSH for 240 h. Follicle diameter and steroid production were assessed every 48 h in both studies. Follicles encapsulated in the 0.5% alginate grew faster (P<0.05) than those cultured in the 1.5% concentration. Oestradiol (E(2)) and progesterone (P(4)) increased consistently (P<0.05) over time, and follicles in the 1.5% alginate produced more (P<0.05) P(4) than those in the 0.5% solution. Follicles cultured in the highest FSH concentration (100 µg/ml) increased 100% in size after 240 h compared with 50 to 70% in lower dosages. E(2) concentration remained unchanged over time (P>0.05) across FSH dosages. However, P(4) increased (P<0.05) as culture progressed and with increasing FSH concentration. Results demonstrate that dog follicles cultured in alginate retain structural integrity, grow in size and are hormonally active. Lower alginate and increasing FSH concentrations promote in vitro follicle growth. However, the absence of an E(2) rise in follicles cultured in FSH alone suggests the need for LH supplementation to support theca cell differentiation and granulosa cell function.


Assuntos
Cães , Estradiol/metabolismo , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/metabolismo , Progesterona/metabolismo , Técnicas de Cultura de Tecidos/veterinária , Alginatos/química , Alginatos/metabolismo , Animais , Sobrevivência Celular , Fenômenos Químicos , Gonadotropina Coriônica/metabolismo , Feminino , Hormônio Foliculoestimulante/metabolismo , Ácido Glucurônico/química , Ácido Glucurônico/metabolismo , Ácidos Hexurônicos/química , Ácidos Hexurônicos/metabolismo , Hidrogel de Polietilenoglicol-Dimetacrilato , Cinética , Hormônio Luteinizante/metabolismo , Concentração Osmolar , Folículo Ovariano/citologia , Técnicas Reprodutivas/veterinária
13.
Science ; 239(4845): 1296-9, 1988 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-3125611

RESUMO

The alterations in morphology and function of the ovarian follicle as it matures, ovulates, and becomes a corpus luteum are dramatic. A variety of steroid and polypeptide hormones influence these processes, and the ovary in turn produces specific hormonal signals for endocrine regulation. One such signal is inhibin, a heterodimeric protein that suppresses the secretion of follicle-stimulating hormone from pituitary gonadotrophs. Rat inhibin complementary DNA probes have been used to examine the levels and distribution of inhibin alpha-and beta A-subunit messenger RNAs in the ovaries of cycling animals. Striking, dynamic changes have been found in inhibin messenger RNA accumulation during the developmental maturation of the ovarian follicle.


Assuntos
Estro , Inibinas/genética , Folículo Ovariano/fisiologia , RNA Mensageiro/genética , Animais , Feminino , Hormônio Foliculoestimulante/sangue , Hormônio Luteinizante/sangue , Substâncias Macromoleculares , Hibridização de Ácido Nucleico , Ovário/fisiologia , RNA Mensageiro/metabolismo , Ratos
14.
Mol Cell Biol ; 6(8): 2932-43, 1986 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-3537727

RESUMO

We identified and produced antibodies to the major proteins that interact with poly(A)+ RNAs in the yeast Saccharomyces cerevisiae. The major proteins which were cross-linked by UV light to poly(A)+ RNA in intact yeast cells had apparent molecular weights of 72,000, 60,000, and 50,000. The poly(A) segment of the RNA was selectively cross-linked to the 72,000-molecular-weight protein (72K protein). Mice immunized with purified UV-cross-linked RNA-protein (RNP) complexes produced antibodies to the three major RNP proteins. A yeast genomic DNA library constructed in the lambda gt11 expression vector was screened with the anti-RNP serum, and recombinant bacteriophage clones were isolated. One recombinant phage, lambda YPA72.1, bearing a 2.5-kilobase insert, produced a large beta-galactosidase-RNP fusion protein. Affinity-selected antibodies from the anti-RNP serum on this fusion protein recognized a single 72K protein which was cross-linked to the poly(A) segment of RNA in the intact cell. Furthermore, the fusion protein of lambda YPA72.1 had specific poly(A)-binding activity. Therefore, lambda YPA72.1 encodes the 72K poly(A)-binding protein. Immunofluorescence microscopy showed that this protein was localized in the cytoplasm. Hybrid-selected mRNA translated in vitro produced the 72K poly(A)-binding protein, and mRNA blot analysis detected a single 2.1-kilobase mRNA. DNA blot analysis suggested a single gene for the poly(A)-binding protein. DNA sequence analysis of genomic clones spanning the entire gene revealed a long open reading frame encoding a 64,272-molecular-weight protein with several distinct domains and repeating structural elements. A sequence of 11 to 13 amino acids is repeated three times in this protein. Strikingly, this repeated sequence (RNP consensus sequence) is highly homologous to a sequence that is repeated twice in a major mammalian heterogeneous nuclear RNP protein, A1. The conservation of the repetitive RNP consensus sequence suggests an important function and a common evolutionary origin for messenger RNP and heterogeneous nuclear RNP proteins.


Assuntos
Proteínas de Transporte/genética , RNA Mensageiro/metabolismo , Ribonucleoproteínas/isolamento & purificação , Sequência de Aminoácidos , Sequência de Bases , Técnicas de Imunoadsorção , Microscopia de Fluorescência , Hibridização de Ácido Nucleico , Poli A/metabolismo , Proteínas de Ligação a Poli(A) , Biossíntese de Proteínas , Saccharomyces cerevisiae/genética
15.
Trends Endocrinol Metab ; 11(8): 309-14, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10996525

RESUMO

Many of the signal transduction pathways required for mammalian endocrine cell function are conserved from flies and worms. These model organisms permitted the illumination of the biological properties of ligands and provided systems in which cellular coactivating molecules could be identified rapidly. Our knowledge about the activin signaling components has been advanced tremendously by the work carried out in these systems. Subsequent research is beginning to reveal the complex interactions that serve to regulate this signaling pathway.


Assuntos
Substâncias de Crescimento/genética , Substâncias de Crescimento/fisiologia , Inibinas/genética , Inibinas/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Ativinas , Animais , Humanos , Inibinas/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos
16.
Mol Endocrinol ; 15(4): 654-67, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11266515

RESUMO

Inhibin binding protein (InhBP) and the transforming growth factor-beta (TGF beta) type III receptor, beta glycan, have been identified as putative inhibin coreceptors. Here we cloned the InhBP cDNA in rats and predict that it encodes a large membrane-spanning protein that is part of the Ig superfamily, as has been described for humans. Two abundant InhBP transcripts (4.4 and 1.8 kb) were detected in the adult rat pituitary. The larger transcript encodes the full-length protein while the 1.8-kb transcript (InhBP-short or InhBP-S) corresponds to a splice variant of the receptor. This truncated isoform contains only the N-terminal signal peptide and first two (of 12) Ig-like domains observed in the full-length InhBP (InhBP-long or InhBP-L). InhBP-S does not contain a transmembrane domain and is predicted to be a soluble protein. Beta glycan was also detected in the pituitary; however, it was most abundant within the intermediate lobe. Although we also observed beta glycan immunopositive cells in the anterior pituitary, they rarely colocalized with FSH beta-producing cells. We next examined physiological regulation of the coreceptors across the rat estrous cycle. Like circulating inhibin A and inhibin B levels, pituitary InhBP-L and InhBP-S mRNA levels were dynamically regulated across the cycle and were negatively correlated with serum FSH levels. Expression of both forms of InhBP was also positively correlated with serum inhibin B, but not inhibin A, levels. These data are particularly interesting in light of our in vitro observations that InhBP may function as an inhibin B-specific coreceptor. Pituitary beta glycan mRNA levels did not fluctuate across the cycle nor did they correlate with serum FSH. These observations, coupled with its pattern of expression within the pituitary, indicate that beta glycan likely functions as more than merely an inhibin coreceptor within the pituitary. A direct role for InhBP or beta glycan in regulation of pituitary FSH by inhibin in vivo has yet to be determined, but the demonstration of dynamic regulation of pituitary InhBP and its negative relation to serum FSH across the estrous cycle is an important step in this direction.


Assuntos
Processamento Alternativo , Estro/fisiologia , Hipófise/fisiologia , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Receptores de Ativinas , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Feminino , Hormônio Foliculoestimulante/sangue , Regulação da Expressão Gênica , Humanos , Inibinas/metabolismo , Masculino , Dados de Sequência Molecular , Proteoglicanas/genética , Ratos , Ratos Sprague-Dawley , Receptores de Fatores de Crescimento Transformadores beta/genética , Testículo/fisiologia , Transcrição Gênica
17.
Mol Endocrinol ; 15(4): 668-79, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11266516

RESUMO

An antagonistic relationship between inhibin and activin is essential to the control of pituitary FSH release and to normal gonadal function. Two inhibin ligands, inhibin A and inhibin B, are made by the ovary in females, and each regulate pituitary FSH at different times during the reproductive cycle. Inhibin B, but not inhibin A, is produced by the testes and is therefore responsible for all inhibin-dependent FSH regulation in males. Although the activin signal transduction pathway has been well characterized, little is known about the mechanism of inhibin signaling and its relationship to activin antagonism. A recently cloned inhibin-binding protein, InhBP (p120), associates strongly with the type IB activin receptor (Alk4) in a ligand-responsive manner and interacts to a lesser extent with other activin and bone morphogenetic protein (BMP) type I and activin type II receptors. Activin stimulates the association of InhBP and Alk4, and inhibin B, but not inhibin A, interferes with InhBP-Alk4 complex formation. InhBP is necessary to mediate a specific antagonistic effect of inhibin B on activin-stimulated transcription. Appropriate stoichiometry between InhBP and the activin type I receptor is crucial to InhBP function. These findings suggest that InhBP is an inhibin B-specific receptor that mediates antagonism of activin signal transduction through the modulation of activin heteromeric receptor complex assembly.


Assuntos
Inibinas/metabolismo , Proteínas Secretadas pela Próstata , Receptores de Peptídeos/metabolismo , Receptores de Ativinas , Receptores de Ativinas Tipo I , Ativinas , Animais , Células Cultivadas , Inibinas/genética , Receptores de Fatores de Crescimento/metabolismo , Receptores de Peptídeos/genética , Transdução de Sinais , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo
18.
Mol Endocrinol ; 1(8): 561-8, 1987 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-3153478

RESUMO

Inhibin is a gonadal protein hormone that suppresses the secretion of FSH from pituitary gonadotrophs. It has previously been characterized as a heterodimer of two dissimilar subunits (alpha, 18 kilodaltons and beta, 14 kilodaltons) the smaller of which exists in two forms (beta A and beta B) and can form dimers that stimulate the secretion of FSH. In the present work, cDNA clones encoding the inhibin alpha- and beta A-subunits have been isolated from rat ovary and characterized. The alpha-inhibin cDNA predicts a precursor protein of 366 amino acids containing the 133 amino acid mature alpha-subunit at its COOH-terminus. The beta A-inhibin cDNA predicts a precursor protein of 424 amino acids containing the 116 amino acid beta A-subunit at its COOH-terminus. Analysis of rat ovarian RNA indicates that alpha-inhibin mRNA levels are stimulated by PMSG treatment in vivo. In cultured granulosa cells, FSH also stimulates alpha-inhibin mRNA, and the FSH effect is suppressed by cotreatment with GnRH. Hybridization in situ to rat ovarian tissue demonstrates that both the alpha-inhibin and beta A-inhibin mRNAs are specifically expressed in granulosa cells of the developing follicles.


Assuntos
Células da Granulosa/metabolismo , Inibinas/genética , Fragmentos de Peptídeos/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Células Cultivadas , Clonagem Molecular , DNA/genética , Feminino , Dados de Sequência Molecular , RNA Mensageiro/biossíntese , Ratos , Ratos Endogâmicos
19.
Mol Endocrinol ; 10(5): 534-43, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8732684

RESUMO

Activins and inhibins, members of the transforming growth factor-beta superfamily, are involved in diverse physiological and developmental processes. We have previously shown that mice deficient in alpha-inhibin develop gonadal sex cord-stromal tumors at an early age. The tumor development is rapidly followed by a wasting syndrome that includes severe weight loss, hepatocellular necrosis around the central vein, and depletion of the parietal cells in the glandular stomach. The liver histology in inhibin-deficient mice is similar to the pathological effects of short-term treatment of rats and mice with recombinant activin A. Consistent with these findings, we have shown that the gonadal tumors in the inhibin-deficient mice secrete high levels of activins. In addition, Northern blot analysis has localized activin receptor type II (ActRII) to the liver. Based on these studies, we postulated that tumor-produced activins act through ActRII to cause the wasting syndrome in inhibin-deficient mice. To test this hypothesis and determine the significance of elevated levels of activin signaling through ActRII in vivo, we generated compound homozygous mutant mice deficient in both alpha-inhibin and ActRII. Despite the continued development of gonadal sex cord-stromal tumors and elevated serum levels of activin A and B, the compound homozygous mutant mice suffered no unusual weight loss, and the stomachs and livers of the majority of the mice were histologically normal. These results demonstrate that increased levels of activin signaling through ActRII in hepatocytes and the glandular stomach causes the hepatocellular necrosis and depletion of parietal cells in the glandular stomach as well as the severe weight loss in vivo.


Assuntos
Caquexia/etiologia , Inibinas/deficiência , Inibinas/metabolismo , Oligopeptídeos , Receptores de Fatores de Crescimento/fisiologia , Transdução de Sinais , Receptores de Ativinas , Ativinas , Animais , Feminino , Fígado/patologia , Masculino , Camundongos , Camundongos Mutantes , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Peptídeos/metabolismo , Tumores do Estroma Gonadal e dos Cordões Sexuais/metabolismo , Tumores do Estroma Gonadal e dos Cordões Sexuais/patologia , Estômago/patologia , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/patologia , Redução de Peso
20.
Mol Endocrinol ; 13(6): 851-65, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10379885

RESUMO

The role of FSH in gonadal tumorigenesis and, in particular, in human ovarian cancer has been debated. It is also unclear what role the elevated FSH levels in the inhibin-deficient mouse play in the gonadal tumorigenesis. To directly assess the role of FSH in gonadal growth, differentiation, and gonadal tumorigenesis, we have generated both gain-of-function and loss-of-function transgenic mutant mice. In the gain-of-function model, we have generated transgenic mice that ectopically overexpress human FSH from multiple tissues using a mouse metallothionein-1 promoter, achieving levels far exceeding those seen in postmenopausal women. Male transgenic mice are infertile despite normal testicular development and demonstrate enlarged seminal vesicles secondary to elevated serum testosterone levels. Female transgenic mice develop highly hemorrhagic and cystic ovaries, have elevated serum estradiol and progesterone levels, and are infertile, mimicking the features of human ovarian hyperstimulation and polycystic ovarian syndromes. Furthermore, the female transgenic mice develop enlarged and cystic kidneys and die between 6-13 weeks as a result of urinary bladder obstruction. In a complementary loss-of-function approach, we have generated double-homozygous mutant mice that lack both inhibin and FSH by a genetic intercross. In contrast to male mice lacking inhibin alone, 95% of which die of a cancer cachexia-like syndrome by 12 weeks of age, only 30% of the double-mutant male mice lacking both FSH and inhibin die by 1 yr of age. The remaining double-mutant male mice develop slow-growing and less hemorrhagic testicular tumors, which are noted after 12 weeks of age, and have minimal cachexia. Similarly, the double-mutant female mice develop slow-growing, less hemorrhagic ovarian tumors, and 70% of these mice live beyond 17 weeks. The double-mutant mice demonstrate minimal cachexia in contrast to female mice lacking only inhibin, which develop highly hemorrhagic ovarian tumors, leading to cachexia and death by 17 weeks of age in 95% of the cases. The milder cachexia-like symptoms of the inhibin and FSH double-mutant mice are correlated with low levels of serum estradiol and activin A and reduced levels of aromatase mRNA in the gonadal tumors. Based on these and our previous genetic analyses, we conclude that elevated FSH levels do not directly cause gonadal tumors. However, these results suggest FSH is an important trophic modifier factor for gonadal tumorigenesis in inhibin-deficient mice.


Assuntos
Hormônio Foliculoestimulante/genética , Inibinas/genética , Oligopeptídeos , Neoplasias Ovarianas/genética , Ovário/crescimento & desenvolvimento , Neoplasias Testiculares/genética , Ativinas , Animais , Cruzamentos Genéticos , Feminino , Hormônio Foliculoestimulante/metabolismo , Regulação da Expressão Gênica , Hemorragia/genética , Homozigoto , Humanos , Infertilidade Feminina/genética , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Inibinas/sangue , Inibinas/metabolismo , Masculino , Metalotioneína/genética , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Neoplasias Ovarianas/patologia , Ovário/patologia , Peptídeos/sangue , Peptídeos/genética , Síndrome do Ovário Policístico/genética , Glândulas Seminais/patologia , Esteroides/sangue , Neoplasias Testiculares/patologia , Sistema Urinário/anormalidades , Síndrome de Emaciação/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA