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.
Reprod Biomed Online ; 48(2): 103695, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38177037

RESUMO

Similar to diabetes and unlike many pathogen-induced diseases, endometriosis is likely a result of maladaptation to the evolutionary heritage of humans. The objective of this article is to review the literature and improve understanding of the evolutionary factors behind endometriosis, leading to more effective prevention and treatment approaches. In primates, spontaneous decidualization of the endometrium evolved to ensure optimal implantation of a limited number of early embryos, unlike many non-primates which depend on early embryos to induce decidualization and subsequent pregnancy. Spontaneous decidualization results in menstrual bleeding when embryo implantation does not occur, and endometriosis is commonly believed to be caused by retrograde menstruation. Although direct evidence is lacking, it is likely that hunter-gatherer women experienced fewer menstrual periods due to pregnancy shortly after menarche, followed by repeated pregnancies and lactation. However, the mismatch between the evolved uterine physiology and rapid societal changes has led to modern women delaying pregnancy and experiencing numerous menstrual periods, potentially increasing the incidence of endometriosis. The symptoms of endometriosis are often managed by suppressing menstruation through systemic hormonal treatments, but these may have side effects. For patients with a family history of endometriosis or in the early stages of the disease, intrauterine devices releasing progesterone locally could prevent uterine bleeding and the development of endometriosis while preserving fertility and minimizing side effects.


Assuntos
Endometriose , Gravidez , Animais , Feminino , Humanos , Endometriose/complicações , Progesterona , Menstruação , Hemorragia Uterina , Endométrio/fisiologia
2.
Hum Reprod ; 37(9): 1959-1969, 2022 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-35881063

RESUMO

During human evolution, major changes in our societal conditions and environment took place without sufficient time for concomitant genetic alterations, leading to out of step adaptation and diseases in women. We first discuss recent societal adaptation mismatch (menstrual bleeding; increases in cancers of reproductive organs, endometriosis; mother's nursing; polycystic ovarian syndrome; transgenerational epigenetic modifications), followed by Darwinian out of step adaptation (labor difficulties; sex chromosomes, human diseases and sex disparity in genomic DNA). We discuss the evolutionary basis of menstrual bleeding, followed by recent increases in cancers of reproductive organs and endometriosis. The importance of breastfeeding by mothers is also emphasized. Earlier onset of menarche, decreased rates of childbirths and breastfeeding resulted in increased number of menstrual cycles in a lifetime, coupled with excess estrogen exposure and incessant ovulation, conditions that increased the susceptibility to mammary and uterine cancers as well as ovarian epithelial cancer and endometriosis. Shorter lactation duration in mothers also contributed to more menstrual cycles. We further discuss the evolutionary basis of the prevalent polycystic ovary syndrome. During the long-term Darwinian evolution, difficulties in childbirth evolved due to a narrowed pelvis, our upright walking and enlarged fetal brain sizes. Because there are 1.5% genomic DNA differences between woman and man, it is of significance to investigate sex-specific human physiology and diseases. In conclusion, understanding out of step adaptation during evolution could allow the prevention and better management of female reproductive dysfunction and diseases.


Assuntos
Endometriose , Síndrome do Ovário Policístico , Endometriose/genética , Feminino , Humanos , Masculino , Ciclo Menstrual/fisiologia , Menstruação , Saúde da Mulher
3.
FASEB J ; 34(7): 8824-8832, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32501617

RESUMO

Sequencing diverse genomes allowed the tracing of orthologous and paralogous genes to understand the co-evolution of polypeptide ligands and receptors. This review documents the discovery of several polypeptide ligands and their cognate receptors mainly expressed in the reproductive tissue using evolutionary genomics. We discussed the sub-functionization of paralogs and co-evolution of ligand-receptor families. Based on the conserved signaling among paralogous receptors and common knock-out phenotypes of ligand-receptor pairs, relationships between relaxin family peptides and leucine-rich repeat-containing, G protein-coupled receptors (LGR) were revealed. We also described the identification of a novel paralogous glycoprotein hormone thyrostimulin and design of a long-acting FSH. Human stresscopin and stresscopin-related peptide, paralogous to CRH, were also identified based on the conserved signaling pathways. Recently, a novel ligand placensin expressed in human placenta was found based on the paralogous relationship with a metabolic hormone asprosin. Placensin likely contributes to stage-dependent increases in insulin resistance during human pregnancy and its elevated secretion was associated with gestational diabetes mellitus. Although many ligands were predicted based on sequence signatures, ligands of shorter sequences have not been identified, together with many "orphan" receptors without known ligands. Future development of tools for predicting ligands and high throughput assays to identify ligand-receptor pairs based on ligand binding and/or signal transduction could advance hormone-based physiology and pathophysiology.


Assuntos
Evolução Molecular , Fragmentos de Peptídeos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Animais , Humanos , Ligantes
4.
Reprod Biomed Online ; 40(2): 245-253, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31753712

RESUMO

RESEARCH QUESTION: The recently developed in-vitro activation (IVA) approach provides a promising infertility treatment for patients with premature ovarian insufficiency. The IVA method promotes growth of residual ovarian follicles following ovarian tissue fragmentation leading to Hippo signalling disruption, together with in-vitro incubation with Akt stimulators. As poor ovarian response (POR) patients with decreased ovarian reserve (DOR) have multiple secondary follicles, this study tested whether Hippo signalling disruption alone using in-vitro ovarian cortical fragmentation, followed by autologous grafting, was sufficient to promote follicle growth. DESIGN: A case series study. RESULTS: In 9 out of 11 POR patients with DOR treated with a simplified IVA procedure, increases in antral follicle numbers in multiple growth waves were detected following FSH treatment. Subsequent injection with human chorionic gonadotrophin allowed retrieval of more mature oocytes for IVF (median antral follicle counts before and after IVA per ovarian stimulation: 1.0 versus 2.6) with 68.7% fertilization rates and 56.9% showing high-quality embryonic development. One natural conception and 16 embryo transfers in five patients resulted in one live birth, two ongoing pregnancies and one miscarriage. Three additional patients and the miscarriage patient have cryopreserved embryos for future transfer. CONCLUSIONS: The present drug-free IVA approach may be suitable for POR patients with DOR, as it increased the number of antral follicles. The procedure also eliminated the need for 2-day incubation with drugs and required only one surgery. This approach could allow the retrieval of more oocytes in middle-aged women to achieve higher pregnancy rates and deserves proper evaluation in future randomized controlled trials.


Assuntos
Fertilização in vitro/métodos , Infertilidade Feminina/terapia , Folículo Ovariano/fisiologia , Reserva Ovariana/fisiologia , Indução da Ovulação/métodos , Adulto , Hormônio Antimülleriano/sangue , Feminino , Hormônio Foliculoestimulante/sangue , Humanos , Recuperação de Oócitos/métodos , Ovário/fisiologia , Gravidez , Taxa de Gravidez
5.
Reprod Biomed Online ; 36(5): 584-593, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29602728

RESUMO

During each reproductive cycle, the ovary exhibits tissue remodelling and cyclic vasculature changes associated with hormonally regulated folliculogenesis, follicle rupture, luteal formation and regression. However, the relationships among different types of follicles and corpora lutea are unclear, and the role of ovarian vasculature in folliculogenesis and luteal dynamics has not been extensively investigated. Understanding of ovarian physiology and pathophysiology relies upon elucidation of ovarian morphology and architecture. This paper summarizes the literature on traditional approaches to the imaging of ovarian structures and discusses recent advances in ovarian imaging. Traditional in-vivo ultrasound, together with histological and electron microscopic approaches provide detailed views of the ovary at organ, tissue and molecular levels. However, in-vivo imaging is limited to antral and larger follicles whereas histological imaging is mainly two-dimensional in nature. Also discussed are emerging approaches in the use of near-infrared fluorophores to image follicles in live animals to detect preantral follicles as well as visualizing ovarian structures using CLARITY in fixed whole ovaries to elucidate three-dimensional interrelationships among follicles, corpora lutea and ovarian vasculature. Advances in ovarian imaging techniques provide new understanding of ovarian physiology and allow for the development of better tools to diagnose ovarian pathophysiology.


Assuntos
Ovário/diagnóstico por imagem , Animais , Diagnóstico por Imagem/métodos , Diagnóstico por Imagem/tendências , Feminino , Humanos , Camundongos , Doenças Ovarianas/diagnóstico por imagem
6.
J Biol Chem ; 290(30): 18261-8, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-25995451

RESUMO

Apela (APJ early endogenous ligand, also known as elabela or toddler) is a recently discovered peptide hormone. Based on genetic studies in zebrafish, apela was found to be important for endoderm differentiation and heart development during embryogenesis. Although common phenotypes of apela and APJ-null zebrafish during embryonic development suggested that apela interacts with the APJ receptor, kinetics of apela binding to APJ and intracellular signaling pathways for apela remain unknown. The role of apela in adults is also uncertain. Using a chimeric apela ligand, we showed direct binding of apela to APJ with high affinity (Kd = 0.51 nm) and the ability of apelin, the known peptide ligand for APJ, to compete for apela binding. Apela, similar to apelin, acts through the inhibitory G protein pathway by inhibiting forskolin-stimulated cAMP production and by inducing ERK1/2 phosphorylation. In adult rats, apela is expressed exclusively in the kidney, unlike the wide tissue distribution of apelin. In vivo studies demonstrated the ability of apela to regulate fluid homeostasis by increasing diuresis and water intake. Dose-response studies further indicated that apela induces 2- and 5-fold higher maximal responses than apelin in ERK1/2 phosphorylation and diuresis/water intake, respectively. After designing an apela antagonist, we further demonstrated the role of endogenous ligand(s) in regulating APJ-mediated fluid homeostasis. Our results identified apela as a potent peptide hormone capable of regulating fluid homeostasis in adult kidney through coupling to the APJ-mediated Gi signaling pathway.


Assuntos
Proteínas de Ligação ao GTP/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Rim/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Apelina , Células CHO , Cricetulus , Desenvolvimento Embrionário/genética , Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Homeostase/genética , Homeostase/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Rim/fisiologia , Ligantes , Hormônios Peptídicos/genética , Hormônios Peptídicos/metabolismo , Ligação Proteica , Ratos , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/genética , Água/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/biossíntese , Proteínas de Peixe-Zebra/genética
7.
Curr Opin Obstet Gynecol ; 28(3): 217-22, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27022685

RESUMO

PURPOSE OF REVIEW: Premature ovarian failure (POF) is diagnosed by amenorrhea before 40 years of age. Owing to exhaustion of follicles in POF ovaries, egg donation is the only option. Although menstrual cycles cease in POF patients, some of them still contain residual dormant follicles in ovaries. Recently, we developed a new infertility treatment and named it as in-vitro activation (IVA), which enables POF patients to conceive using their own eggs by activation of residual dormant follicles. Here, we summarize data showing the potential of IVA as a new infertility treatment for POF patients. RECENT FINDINGS: Transgenic mouse studies revealed that the stimulation of phosphatidylinositol-3-kinase-AKT-forkhead box O3 pathway activated dormant primordial follicles. In murine and human ovaries, the phosphatase and tensin homolog inhibitors and phosphatidylinositol-3-kinase activators were demonstrated to activate dormant primordial follicles in in-vitro cultures. Subsequent studies showed that ovarian fragmentation suppressed Hippo signaling pathway, leading to ovarian follicle growth. Combining these two methods in an IVA approach followed by ovarian tissue autotransplantation, successful follicle growth, and pregnancies were reported in POF patients. Currently, two healthy babies were delivered, together with two additional pregnancies. SUMMARY: IVA treatment is a potential infertility therapy for POF patients who have residual follicles.


Assuntos
Infertilidade Feminina/terapia , Menopausa Precoce , Oócitos/fisiologia , Folículo Ovariano/fisiopatologia , Insuficiência Ovariana Primária/terapia , Técnicas de Reprodução Assistida , Criopreservação , Feminino , Fertilização , Humanos , Transdução de Sinais
8.
J Cell Sci ; 126(Pt 9): 2060-8, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23444378

RESUMO

Mammalian LGR4, 5 and 6 are seven-transmembrane receptors that are important for diverse physiological processes. These receptors are orthologous to DLGR2, a Drosophila receptor activated by the burs/pburs heterodimer important for morphogenesis. Although recent studies indicated that four R-spondin proteins are cognate ligands for LGR4, 5 and 6 receptors, several BMP antagonists in vertebrates have been postulated to be orthologous to burs and pburs. Using newly available genome sequences, we showed that norrin is a vertebrate ortholog for insect burs and pburs and stimulates Wnt signaling mediated by LGR4, but not by LGR5 and 6, in mammalian cells. Although norrin could only activate LGR4, binding studies suggested interactions between norrin and LGR4, 5 and 6. Norrin, the Norrie disease gene product, is also capable of activating Wnt signaling mediated by the Frizzled4 receptor and serves as a BMP antagonist. Mutagenesis studies indicated that different norrin mutations found in patients with Norrie disease can be categorized into subgroups according to defects for signaling through the three distinct binding proteins. Thus, norrin is a rare ligand capable of binding three receptors/binding proteins that are important for BMP and Wnt signaling pathways.


Assuntos
Proteínas do Olho/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Galinhas , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Proteínas do Olho/genética , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Humanos , Camundongos , Proteínas do Tecido Nervoso/genética , Receptores Acoplados a Proteínas G/genética , Via de Sinalização Wnt/fisiologia , Xenopus
9.
FASEB J ; 27(6): 2175-84, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23407710

RESUMO

R-spondin proteins are adult stem cell growth factors capable of stimulating gut development by activating LGR4, 5, and 6 receptors to promote Wnt signaling. Although multiple Wnt ligands and cognate Frizzled receptors are expressed in the ovary, their physiological roles are unclear. Based on bioinformatic and in situ hybridization analyses, we demonstrated the exclusive expression of R-spondin2 in oocytes of ovarian follicles. In cultured somatic cells from preantral follicles, R-spondin2 treatment (ED50: 3 ng/ml) synergized with Wnt3a to stimulate Wnt signaling. In cultured ovarian explants from prepubertal mice containing preantral follicles, treatment with R-spondin2, similar to follicle stimulating hormone, promoted the development of primary follicles to the secondary stage. In vivo administration of an R-spondin agonist stimulated the development of primary follicles to the antral stage in both immature mice and gonadotropin releasing hormone antagonist-treated adult mice. Subsequent treatment with gonadotropins allowed the generation of mature oocytes capable of undergoing early embryonic development and successful pregnancy. Furthermore, R-spondin agonist treatment of immune-deficient mice grafted with human cortical fragments stimulated the development of primary follicles to the secondary stage. Thus, oocyte-derived R-spondin2 is a paracrine factor essential for primary follicle development, and R-spondin agonists could provide a new treatment regimen for infertile women with low responses to the traditional gonadotropin therapy.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Oócitos/metabolismo , Folículo Ovariano/fisiologia , Trombospondinas/fisiologia , Animais , Células Cultivadas , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/agonistas , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos ICR , Camundongos SCID , Folículo Ovariano/citologia , Folículo Ovariano/transplante , Comunicação Parácrina , Gravidez , Trombospondinas/agonistas , Trombospondinas/genética , Transplante Heterólogo
10.
Proc Natl Acad Sci U S A ; 107(22): 10280-4, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20479243

RESUMO

Although multiple follicles are present in mammalian ovaries, most of them remain dormant for years or decades. During reproductive life, some follicles are activated for development. Genetically modified mouse models with oocyte-specific deletion of genes in the PTEN-PI3K-Akt-Foxo3 pathway exhibited premature activation of all dormant follicles. Using an inhibitor of the Phosphatase with TENsin homology deleted in chromosome 10 (PTEN) phosphatase and a PI3K activating peptide, we found that short-term treatment of neonatal mouse ovaries increased nuclear exclusion of Foxo3 in primordial oocytes. After transplantation under kidney capsules of ovariectomized hosts, treated follicles developed to the preovulatory stage with mature eggs displaying normal epigenetic changes of imprinted genes. After in vitro fertilization and embryo transfer, healthy progeny with proven fertility were delivered. Human ovarian cortical fragments from cancer patients were also treated with the PTEN inhibitor. After xeno-transplantation to immune-deficient mice for 6 months, primordial follicles developed to the preovulatory stage with oocytes capable of undergoing nuclear maturation. Major differences between male and female mammals are unlimited number of sperm and paucity of mature oocytes. Thus, short-term in vitro activation of dormant ovarian follicles after stimulation of the PI3K-Akt pathway allows the generation of a large supply of mature female germ cells for future treatment of infertile women with a diminishing ovarian reserve and for cancer patients with cryo-preserved ovaries. Generation of a large number of human oocytes also facilitates future derivation of embryonic stem cells for regenerative medicine.


Assuntos
Oócitos/fisiologia , Folículo Ovariano/citologia , Folículo Ovariano/fisiologia , Animais , Animais Recém-Nascidos , Transferência Embrionária , Feminino , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Humanos , Técnicas In Vitro , Masculino , Camundongos , Oócitos/citologia , Oócitos/efeitos dos fármacos , Compostos Organometálicos/farmacologia , Folículo Ovariano/efeitos dos fármacos , Ovário/transplante , PTEN Fosfo-Hidrolase/antagonistas & inibidores , Fosfatidilinositol 3-Quinases/metabolismo , Gravidez , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transplante Heterólogo
11.
iScience ; 26(10): 108065, 2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37860761

RESUMO

Fish require abundant nutrients to generate a large number of eggs for spawning. Based on the evolutionary conservation of human FBN2 and its C-terminal placensin-like sequences in fish, we identified a peptide hormone gonacin (GONAdal Cell placensIN) and found its high expression in early-stage germ cells in the ovary and testis of zebrafish. We demonstrated that gonacin is essential for food intake, glucose release, and ovarian development in zebrafish. Similar expression patterns and functions of gonacin were also demonstrated in rainbow trout. Gonacin represents the first hormone secreted by germ cells with endocrine functions in vertebrates, bridging the energy homeostasis and reproduction.

12.
Biol Reprod ; 84(4): 707-14, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21178173

RESUMO

Development of ovarian follicles is regulated by pituitary-derived gonadotropins together with local ovarian paracrine factors. Based on DNA microarray data, we performed RT-PCR and immunostaining to demonstrate the expression of interleukin 7 transcripts in oocytes of preantral, antral, and preovulatory follicles in rats. We also found the expression of interleukin 7 receptor and the coreceptor interleukin 2 receptor gamma in granulosa cells, cumulus cells, and preovulatory oocytes. In cultured rat granulosa cells obtained from early antral and preovulatory follicles, treatment with interleukin 7 stimulated the phosphorylation of AKT, glycogen synthase kinase (GSK3B), and STAT5 proteins in a time- and dose-dependent manner. Furthermore, measurement of mitochondrial reductase activity indicated that treatment with interleukin 7, similar to gonadotropins, increased the number of viable granulosa cells during a 24-h culture period. Furthermore, monitoring of the activities of apoptotic enzymes (caspase 3/7) indicated that treatment with interleukin 7 suppressed apoptosis of cultured granulosa cells from both antral and preovulatory follicles following serum withdrawal. The apoptosis-suppressing actions of interleukin 7 were blocked by an inhibitor of the phosphoinositol-3-kinase (PIK3)/AKT pathway. Furthermore, treatment of cultured preovulatory follicles with interleukin 7, like treatment with human chorionic gonadotropin, induced germinal vesicle breakdown of oocytes. The stimulatory effect of interleukin 7 was also blocked by inhibitors of the PIK3/AKT pathway. The present findings suggest that oocyte-derived interleukin 7 could act on neighboring granulosa cells as a survival factor and promote the nuclear maturation of preovulatory oocytes through activation of the PIK3/AKT pathway.


Assuntos
Células da Granulosa/citologia , Células da Granulosa/fisiologia , Interleucina-7/genética , Interleucina-7/fisiologia , Oócitos/crescimento & desenvolvimento , Oócitos/fisiologia , Animais , Apoptose/efeitos dos fármacos , Sequência de Bases , Células Cultivadas , Primers do DNA/genética , Feminino , Expressão Gênica , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Células da Granulosa/efeitos dos fármacos , Subunidade gama Comum de Receptores de Interleucina/genética , Interleucina-7/farmacologia , Oócitos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Interleucina-7/genética , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais
13.
Hum Reprod ; 26(3): 695-702, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21227937

RESUMO

BACKGROUND: In mammalian ovaries, diverse paracrine factors have been identified to mediate or modulate LH-induced changes during ovulation. Due to the difficulty in obtaining non-stimulated granulosa cells during IVF, little is known about the LH-induced paracrine factors in the human ovary. Based on earlier studies using murine ovarian cells showing the paracrine roles of brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) in promoting oocyte maturation, we investigated the expression of these ligands in human granulosa cells and their regulation of human oocyte development. METHODS: Non-stimulated granulosa cells were obtained from non-stimulated IVM (in vitro maturation) patients after oocyte retrieval. Women undergoing non-stimulated IVM treatment at a mean age of 30.8 ± 1.3 (n = 10) were recruited for this study. Immature oocytes and granulosa cells were collected from IVF patients undergoing gonadotrophin stimulation and ICSI. Immunocytochemical analyses of granulosa cells were carried out to investigate expression profiles of BDNF and GDNF, together with real-time RT-PCR to analyze the gonadotrophin regulation of BDNF and GDNF transcript levels. In addition, immature oocytes were cultured to analyze the regulation of oocyte maturation by BDNF and GDNF. RESULTS: BDNF and GDNF were found to be expressed in non-stimulated granulosa cells. After gonadotrophin (FSH and/or hCG) treatment, transcripts levels for BDNF and GDNF were significantly increased (P < 0.05). In cultured immature oocytes, treatment with BDNF or GDNF promoted total yields of metaphase II oocytes. CONCLUSIONS: These findings demonstrate that FSH and hCG treatments augment the expression of BDNF and GDNF by granulosa cells and that these granulosa-cell-derived factors are candidate paracrine factors capable of promoting oocyte maturation.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Gonadotropinas/metabolismo , Células da Granulosa/metabolismo , Oócitos/metabolismo , Oogênese , Adulto , Fator Neurotrófico Derivado do Encéfalo/genética , Divisão Celular , Células Cultivadas , Gonadotropina Coriônica/metabolismo , Feminino , Hormônio Foliculoestimulante/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Células da Granulosa/citologia , Humanos , Infertilidade/terapia , Hormônio Luteinizante/metabolismo , Metáfase/efeitos dos fármacos , Recuperação de Oócitos , Oócitos/citologia , RNA Mensageiro/metabolismo , Injeções de Esperma Intracitoplásmicas
14.
Hum Reprod ; 26(11): 3094-101, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21865234

RESUMO

BACKGROUND: In mammalian follicles, oocytes are arrested at the diplotene stage of prophase I until meiotic resumption following the LH surge. Recently, C-type natriuretic peptide (CNP), encoded by natriuretic peptide precursor type C (NPPC) was found to suppress mouse oocyte maturation by promoting cyclic guanosine 5'-monophospate (cGMP) production in cumulus cells. However, regulation of NPPC/CNP expression during the pre-ovulatory period and their regulation by the LH surge have not been investigated. METHODS AND RESULTS: Based on genome-wide analysis of DNA microarray data sets using samples from periovulatory ovaries, we found increases in NPPC transcripts in granulosa cells during pre-ovulatory follicle growth in mice and a rapid decline induced by the pre-ovulatory LH/hCG stimulation. Treatment of pre-ovulatory animals with hCG decreased ovarian CNP content. In isolated ovarian cells, NPPC mRNA was predominantly expressed in mural granulosa cells exhibiting similar regulation following gonadotrophin treatment. In cultured mouse pre-ovulatory follicles, meiosis resumption in oocytes by hCG treatment was accompanied by decreases in NPPC transcript levels. In cultured mouse cumulus cell-oocyte complexes, CNP treatment inhibited the resumption of meiosis with increases in cGMP levels in both cumulus cells and oocytes. In human ovaries, CNP levels in ovarian follicular fluid were also decreased following treatment of patients with an ovulatory dose of hCG. CONCLUSIONS: Our findings demonstrate gonadotrophins regulation of NPPC/CNP expression in mouse and human ovaries and confirm the role of CNP as a potent paracrine oocyte maturation inhibitor.


Assuntos
Gonadotropina Coriônica/metabolismo , Células da Granulosa/citologia , Hormônio Luteinizante/metabolismo , Meiose , Peptídeo Natriurético Tipo C/metabolismo , Oócitos/citologia , Ovário/metabolismo , Ovulação , Adulto , Animais , Células do Cúmulo/citologia , Feminino , Humanos , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Folículo Ovariano/metabolismo
15.
Comput Struct Biotechnol J ; 19: 2236-2245, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33995916

RESUMO

Acupuncture has been used for treating various medical conditions in traditional Chinese medicine. Both manual and electro-acupuncture stimulate specific acupoints to obtain local and systemic biological effects, but the underlying mechanisms remain unclear. Here, we used three-dimensional tissue-clearing technology to study acupoints on the Ren meridian of mice to reveal the distribution, density, branching, and relationships between blood vessels and nerves. Using topological Mapper methods, we found that sympathetic neurovascular networks were denser in the CV 4 acupoint compared with surrounding non-acupoints. Furthermore, high resolution in vivo real-time vascular imaging using the near infrared-II probe LZ-1105 demonstrated increased blood flow in the CV 4 acupoint compared with neighboring non-acupoints after manual or electro-acupuncture. Consistent with earlier findings, our research indicated that acupuncture could enhance local blood flow, and our high-resolution 3D images show for the first time the important role of sympathetic neurovascular networks in the CV 4 acupoint.

16.
Dev Biol ; 327(1): 62-70, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19111534

RESUMO

Mammalian oocytes remain dormant in the diplotene stage of prophase I until the resumption of meiosis characterized by germinal vesicle breakdown (GVBD) following the preovulatory gonadotropin stimulation. Based on genome-wide analysis of peri-ovulatory DNA microarray to identify paracrine hormone-receptor pairs, we found increases in ovarian transcripts for endothelin-1 and endothelin receptor type A (EDNRA) in response to the preovulatory luteinizing hormone (LH)/human chorionic gonadotropin (hCG) stimulation. Immunohistochemical analyses demonstrated localization of EDNRA in granulosa and cumulus cells. In cultured preovulatory follicles, treatment with endothelin-1 promoted oocyte GVBD. The stimulatory effect of endothelin-1 was blocked by cotreatment with antagonists for the type A, but not related type B, receptor. The stimulatory effect of hCG on GVBD was partially blocked by the same antagonist. The endothelin-1 promotion of GVBD was found to be mediated by the MAPK/ERK pathway but not by the inhibitory G protein. Studies using cumulus-oocyte complexes and denuded oocytes demonstrated that the endothelin-1 actions are mediated by cumulus cells. Furthermore, intrabursal administration with endothelin-1 induced oocyte GVBD in preovulatory follicles. Our findings demonstrate a paracrine role of endothelin-1 in the induction of the resumption of meiosis and provide further understanding on the molecular mechanisms underlying the nuclear maturation of oocytes induced by the preovulatory LH surge.


Assuntos
Endotelina-1/fisiologia , Meiose , Oócitos/citologia , Comunicação Parácrina/fisiologia , Animais , Gonadotropina Coriônica/fisiologia , Células do Cúmulo , Feminino , Hormônio Luteinizante/fisiologia , Camundongos , Folículo Ovariano , Receptores de Endotelina/metabolismo
17.
Fertil Steril ; 114(3): 458-464, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32782158

RESUMO

The Hippo signaling pathway, which is important in organ size regulation, is present in organisms from the fly to mammals. Disruption of the Hippo signaling pathway leads to increased nuclear translocation of the effector Yes-associated protein (YAP), resulting in the expression of cystein-rich 61, connective tissue growth factor, and nephroblastoma overexpressed (CCN) growth factors and baculoviral inhibitors of apoptosis repeat containing (BIRC) apoptosis inhibitors to increase organ sizes. Furthermore, genome-wide knockdown of genes in insect cells demonstrated that actin polymerization promoted nuclear translocation of YAP. In the mammalian ovary, we demonstrated the expression of Hippo signaling pathway genes and showed that ovarian fragmentation increased actin polymerization, leading to YAP nuclear translocation and increased expression of cystein-rich 61, CCN growth factors and BIRC apoptosis inhibitors, followed by enhanced follicle growth. Here we summarize evidence suggesting the role of mechanical stress on follicle growth in the ovary and describe recent use of ovary-damaging procedures to treat ovarian infertility. Ovarian fragmentation, together with in vitro incubation with Akt-stimulating drugs, formed the basis of an in vitro activation (IVA) therapy to treat patients with premature ovarian insufficiency, whereas ovarian fragmentation alone (drug-free IVA) was successful in treating patients with premature ovarian insufficiency with recent menses cessation. For middle-aged women with poor ovarian responses and diminished ovarian reserve, drug-free IVA was also effective in promoting follicle growth for infertility treatment. In addition, an in vivo follicle activation approach based on laparoscopic ovarian incision showed promise for patients with resistant ovary syndrome. With initial success using mechanical disruption approaches, future investigation could evaluate possibilities to refine mechanical methods and to locally administer actin polymerization-enhancing drugs for ovarian infertility treatment.


Assuntos
Infertilidade Feminina/terapia , Folículo Ovariano/metabolismo , Reserva Ovariana , Ovulação , Insuficiência Ovariana Primária/terapia , Proteínas Serina-Treonina Quinases/metabolismo , Técnicas de Reprodução Assistida , Feminino , Fármacos para a Fertilidade Feminina/uso terapêutico , Via de Sinalização Hippo , Humanos , Infertilidade Feminina/metabolismo , Infertilidade Feminina/fisiopatologia , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/fisiopatologia , Reserva Ovariana/efeitos dos fármacos , Ovulação/efeitos dos fármacos , Gravidez , Insuficiência Ovariana Primária/metabolismo , Insuficiência Ovariana Primária/fisiopatologia , Transdução de Sinais , Estresse Mecânico
18.
Dev Biol ; 315(1): 189-202, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18234170

RESUMO

Optimal maturation of oocytes and successful development of preimplantation embryos is essential for reproduction. We performed DNA microarray analyses of ovarian transcripts and identified glial cell line-derived neurotrophic factor (GDNF) secreted by cumulus, granulosa, and theca cells as an ovarian factor stimulated by the preovulatory LH/hCG surge. Treatment of cumulus-oocyte complexes with GDNF enhanced first polar body extrusion with increase in cyclin B1 synthesis and the GDNF actions are likely mediated by its receptor GDNF family receptor-alpha1 (GFRA1) and a co-receptor ret proto-oncogene (Ret), both expressed in oocytes. However, treatment with GDNF did not affect germinal vesicle breakdown and cytoplasmic maturation of oocytes. During the preimplantation stages, GDNF was expressed in pregnant oviducts and uteri, whereas GFRA1 and Ret were expressed in embryos throughout early development with an increase after the early blastocyst stage. In blastocysts, both GDNF and GFRA1 were exclusively localized in trophectoderm cells, whereas Ret was detected in both cell lineages. Treatment with GDNF promoted the development of two-cell-stage embryos into blastocysts showing increased cell proliferation and decreased apoptosis mainly in trophectoderm cells. Our findings suggest potential paracrine roles of GDNF in the promotion of completion of meiosis I and the development of early embryos.


Assuntos
Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Meiose/fisiologia , Oócitos/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Células do Cúmulo/citologia , Ensaio de Imunoadsorção Enzimática , Feminino , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos , Oócitos/citologia , Folículo Ovariano/citologia , Ovário/citologia , Oviductos/metabolismo , Gravidez , Proto-Oncogene Mas , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Estatística como Assunto , Útero/metabolismo
19.
Reproduction ; 138(2): 329-39, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19433502

RESUMO

The expression of hedgehog (Hh) genes, their receptor, and the co-receptor in mice, rat, and bovine ovaries were investigated. RT-PCR of ovarian transcripts in mice showed amplification of transcripts for Indian (Ihh) and desert (Dhh) Hh, patched 1 (Ptch1), and smoothened (Smo) genes. Semi-quantitative RT-PCR and northern blot analyses showed that whole ovarian Ihh and Dhh transcripts decreased 4-24 h after hCG versus 0-48 h after pregnant mares serum gonadotrophin treatment in mice, whereas mouse Ptch1 and Smo transcripts were expressed throughout the gonadotropin treatments. Quantitative real-time RT-PCR (qRT-PCR) revealed that the expression of the Hh-patched signaling system with Ihh mRNA abundance in granulosa cells was greater, whereas Smo and Ptch1 mRNA abundance was less in theca cells of small versus large follicles of cattle. In cultured rat and bovine theca-interstitial cells, qRT-PCR analyses revealed that the abundance of Gli1 and Ptch1 mRNAs were increased (P<0.05) with sonic hedgehog (SHH) treatment. Additional studies using cultured bovine theca cells indicated that SHH induces proliferation and androstenedione production. IGF1 decreased Ihh mRNA abundance in bovine granulosa cells. The expression and regulation of Ihh transcripts in granulosa cells and Ptch1 mRNA in theca cells suggest a potential paracrine role of this system in bovine follicular development. This study illustrates for the first time Hh activation of Gli1 transcriptional factor in theca cells and its stimulation of theca cell proliferation and androgen biosynthesis.


Assuntos
Proteínas Hedgehog/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/fisiologia , Células Tecais/metabolismo , Androstenodiona/biossíntese , Animais , Bovinos , Células Cultivadas , Feminino , Expressão Gênica/efeitos dos fármacos , Gonadotropinas Equinas/farmacologia , Proteínas Hedgehog/análise , Proteínas Hedgehog/genética , Humanos , Insulina/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Hormônio Luteinizante/farmacologia , Camundongos , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Receptores Patched , Receptor Patched-1 , Progesterona/biossíntese , RNA Mensageiro/análise , Ratos , Receptores de Superfície Celular/análise , Receptores de Superfície Celular/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor Smoothened , Transativadores/genética , Transativadores/metabolismo , Proteína GLI1 em Dedos de Zinco
20.
Reprod Biol Endocrinol ; 7: 26, 2009 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-19341483

RESUMO

BACKGROUND: Shortly after stimulation by the preovulatory surge of luteinizing hormone (LH), oocytes arrested at the late prophase I resume meiosis characterized by germinal vesicle breakdown (GVBD), chromosome condensation, and extrusion of the first polar body in preparation for fertilization and early embryonic development. However, oocytes express few or no LH receptors and are insensitive to direct LH stimulation. Thus, factors released by granulosa or theca cells expect to convey the LH stimuli to oocytes. To identify candidate ligand-receptor pairs potentially involved in the process of oocyte maturation, we performed DNA microarray analyses of ovarian transcripts in mice and identified Kit ligand (Kitl) as an ovarian factor stimulated by the LH/hCG surge. The purpose of this study is to investigate the roles of KITL in the nuclear and cytoplasmic maturation of preovulatory mouse oocytes. METHODS: The levels of Kitl and c-kit transcripts in mouse ovaries and isolated ovarian cells were determined by real-time RT-PCR, while expression of KITL protein was examined by immunohistochemistry. Follicle culture, cumulus-oocyte complexes (COC) and denuded oocytes culture were used to evaluate the effect of KITL on mouse oocyte nuclear maturation. To assess the effect of KITL treatment on the cytoplasmic maturation of preovulatory oocytes, we performed in vitro maturation of oocytes followed by in vitro fertilization. RESULTS: Major increase of Kitl transcripts in granulosa cells and mouse ovaries, and predominant expression of c-kit in preovulatory oocytes were identified by real-time RT-PCR. Predominant expression of KITL protein was found in granulosa cells of preovulatory and small antral follicles at 4 h after hCG treatment. In vitro cultures demonstrated that treatment with KITL enhanced first polar body extrusion in a dose-dependent manner. Moreover, treatment of COC with KITL enhanced first polar body extrusion with increase in cyclin B1 synthesis which is important for the progression of meiotic maturation after GVBD. In contrast, treatment of cultured preovulatory follicles with KITL did not affect GVBD and KITL has no effect on cytoplasmic maturation of preovulatory oocytes. CONCLUSION: Our findings suggest potential paracrine roles of KITL in the nuclear maturation of preovulatory oocytes by promoting first polar body extrusion.


Assuntos
Oócitos/metabolismo , Fator de Células-Tronco/fisiologia , Animais , Antígenos/análise , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/fisiologia , Células Cultivadas , Gonadotropina Coriônica/farmacologia , Células do Cúmulo/metabolismo , Citoplasma/efeitos dos fármacos , Citoplasma/fisiologia , Feminino , Fertilização in vitro , Células da Granulosa/metabolismo , Hormônio Luteinizante/farmacologia , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Oócitos/citologia , Oócitos/efeitos dos fármacos , Ovário/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Células-Tronco/metabolismo , Fator de Células-Tronco/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA