Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Res Sq ; 2024 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-38405920

RESUMO

Craniometaphyseal dysplasia (CMD), a rare craniotubular disorder, occurs in an autosomal dominant (AD) or autosomal recessive (AR) form. CMD is characterized by hyperostosis of craniofacial bones and flaring metaphyses of long bones. Many patients with CMD suffer from neurological symptoms. To date, the pathogenesis of CMD is not fully understood. Treatment is limited to decompression surgery. Here, we report a knock in (KI) mouse model for AR CMD carrying a R239Q mutation in CX43. Cx43KI/KI mice replicate many features of AR CMD in craniofacial and long bones. In contrast to Cx43+/+ littermates, Cx43KI/KI mice exhibit periosteal bone deposition and increased osteoclast (OC) numbers in the endosteum of long bones, leading to an expanded bone marrow cavity and increased cortical bone thickness. Although formation of Cx43+/+ and Cx43KI/KI resting OCs are comparable, on bone chips the actively resorbing Cx43KI/KI OCs resorb less bone. Cortical bones of Cx43KI/KI mice have an increase in degenerating osteocytes and empty lacunae. Osteocyte dendrite formation is decreased with reduced expression levels of Fgf23, Sost, Tnf-α, IL-1ß, Esr1, Esr2, and a lower Rankl/Opg ratio. Female Cx43KI/KI mice display a more severe phenotype. Sexual dimorphism in bone becomes more evident as mice age. Our data show that the CX43R239Q mutation results in mislocalization of CX43 protein and impairment of gap junction and hemichannel activity. Different from CX43 ablation mouse models, the CX43R239Q mutation leads to the AR CMD-like phenotype in Cx43KI/KI mice not only by loss-of-function but also via a not yet revealed dominant function.

3.
Endocrinology ; 165(3)2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38180498

RESUMO

Signaling in the granulosa cells of mammalian ovarian follicles is necessary for maintaining prophase arrest in the oocyte and for mediating the resumption of meiosis in response to luteinizing hormone (LH). However, the follicle also includes an outer layer of theca cells, some of which express receptors for LH. To investigate whether theca cells are required for maintaining meiotic arrest and reinitiating meiosis in response to LH, we mechanically separated the granulosa cells and oocyte from the theca and basal lamina. This was accomplished by cutting a slit in the outer surface of isolated follicles such that the mural granulosa cells and cumulus-oocyte complex were extruded from the theca shell, forming a lawn of cells on an organotypic membrane. The remnant of theca cells and basal lamina was then removed. The separation of the granulosa cells from the theca cells and basal lamina was demonstrated by immunofluorescence localization of endomucin (blood vessels of the theca) and laminin gamma (basal lamina). Cells comprising these granulosa cell-oocyte complexes expressed LH receptors and were connected by gap junctions. Oocytes within these granulosa cell complexes maintained meiotic arrest and resumed meiosis in response to LH, showing that the granulosa cells alone, without theca cells, transduce these signals. This semi-intact and mostly 2-dimensional preparation could facilitate imaging studies of follicle physiology.


Assuntos
Hormônio Luteinizante , Células Tecais , Feminino , Animais , Hormônio Luteinizante/farmacologia , Oócitos , Células da Granulosa , Folículo Ovariano , Meiose , Mamíferos
4.
J Immunol ; 212(4): 663-676, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38149920

RESUMO

Implanted medical devices, from artificial heart valves and arthroscopic joints to implantable sensors, often induce a foreign body response (FBR), a form of chronic inflammation resulting from the inflammatory reaction to a persistent foreign stimulus. The FBR is characterized by a subset of multinucleated giant cells (MGCs) formed by macrophage fusion, the foreign body giant cells (FBGCs), accompanied by inflammatory cytokines, matrix deposition, and eventually deleterious fibrotic implant encapsulation. Despite efforts to improve biocompatibility, implant-induced FBR persists, compromising the utility of devices and making efforts to control the FBR imperative for long-term function. Controlling macrophage fusion in FBGC formation presents a logical target to prevent implant failure, but the actual contribution of FBGCs to FBR-induced damage is controversial. CD13 is a molecular scaffold, and in vitro induction of CD13KO bone marrow progenitors generates many more MGCs than the wild type, suggesting that CD13 regulates macrophage fusion. In the mesh implant model of FBR, CD13KO mice produced significantly more peri-implant FBGCs with enhanced TGF-ß expression and increased collagen deposition versus the wild type. Prior to fusion, increased protrusion and microprotrusion formation accompanies hyperfusion in the absence of CD13. Expression of fusogenic proteins driving cell-cell fusion was aberrantly sustained at high levels in CD13KO MGCs, which we show is due to a novel CD13 function, to our knowledge, regulating ubiquitin/proteasomal protein degradation. We propose CD13 as a physiologic brake limiting aberrant macrophage fusion and the FBR, and it may be a novel therapeutic target to improve the success of implanted medical devices. Furthermore, our data directly implicate FBGCs in the detrimental fibrosis that characterizes the FBR.


Assuntos
Corpos Estranhos , Reação a Corpo Estranho , Camundongos , Animais , Reação a Corpo Estranho/induzido quimicamente , Reação a Corpo Estranho/metabolismo , Células Gigantes de Corpo Estranho/metabolismo , Inflamação/metabolismo , Corpos Estranhos/metabolismo , Próteses e Implantes/efeitos adversos , Ubiquitinação
5.
J Cell Biol ; 222(7)2023 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-37102999

RESUMO

Skin homeostasis is maintained by stem cells, which must communicate to balance their regenerative behaviors. Yet, how adult stem cells signal across regenerative tissue remains unknown due to challenges in studying signaling dynamics in live mice. We combined live imaging in the mouse basal stem cell layer with machine learning tools to analyze patterns of Ca2+ signaling. We show that basal cells display dynamic intercellular Ca2+ signaling among local neighborhoods. We find that these Ca2+ signals are coordinated across thousands of cells and that this coordination is an emergent property of the stem cell layer. We demonstrate that G2 cells are required to initiate normal levels of Ca2+ signaling, while connexin43 connects basal cells to orchestrate tissue-wide coordination of Ca2+ signaling. Lastly, we find that Ca2+ signaling drives cell cycle progression, revealing a communication feedback loop. This work provides resolution into how stem cells at different cell cycle stages coordinate tissue-wide signaling during epidermal regeneration.


Assuntos
Sinalização do Cálcio , Cálcio , Pontos de Checagem do Ciclo Celular , Epiderme , Animais , Camundongos , Cálcio/metabolismo , Ciclo Celular , Epiderme/metabolismo
6.
Traffic ; 22(6): 174-179, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33797162

RESUMO

Intercellular organelle transfer has been documented in several cell types and has been proposed to be important for cell-cell communication and cellular repair. However, the mechanisms by which organelle transfer occurs are uncertain. Recent studies indicate that the gap junction protein, connexin 43 (Cx43), is required for mitochondrial transfer but its specific role is unknown. Using three-dimensional electron microscopy and immunogold labeling of Cx43, this report shows that whole organelles including mitochondria and endosomes are incorporated into double-membrane vesicles, called connexosomes or annular gap junctions, that form as a result of gap junction internalization. These findings demonstrate a novel mechanism for intercellular organelle transfer mediated by Cx43 gap junctions.


Assuntos
Endossomos , Junções Comunicantes , Comunicação Celular , Linhagem Celular , Endossomos/metabolismo , Junções Comunicantes/metabolismo , Mitocôndrias
7.
J Cell Sci ; 134(1)2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33277382

RESUMO

Gap junctions have well-established roles in cell-cell communication by way of forming permeable intercellular channels. Less is understood about their internalization, which forms double membrane vesicles containing cytosol and membranes from another cell called connexosomes or annular gap junctions. Here, we systematically investigated the fate of connexosomes in intact ovarian follicles. High-pressure frozen, serial-sectioned tissue was immunogold labeled for connexin 43 (Cx43, also known as GJA1). Within a volume corresponding to ∼35 cells, every labeled structure was categorized and had its surface area measured. Measurements support the concept that multiple connexosomes form from larger invaginated gap junctions. Subsequently, the inner and outer membranes separate, Cx43 immunogenicity is lost from the outer membrane, and the inner membrane appears to undergo fission. One pathway for processing involves lysosomes, based on localization of cathepsin B to some processed connexosomes. In summary, this study demonstrates new technology for high-resolution analyses of gap junction processing.This article has an associated First Person interview with the first author of the paper.


Assuntos
Comunicação Celular , Junções Comunicantes , Feminino , Humanos , Lisossomos , Microscopia Eletrônica , Microscopia Imunoeletrônica , Folículo Ovariano
8.
PLoS Biol ; 16(1): e2003698, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29337984

RESUMO

The Wnt family of secreted proteins has been proposed to play a conserved role in early specification of the bilaterian anteroposterior (A/P) axis. This hypothesis is based predominantly on data from vertebrate embryogenesis as well as planarian regeneration and homeostasis, indicating that canonical Wnt (cWnt) signaling endows cells with positional information along the A/P axis. Outside of these phyla, there is strong support for a conserved role of cWnt signaling in the repression of anterior fates, but little comparative support for a conserved role in promotion of posterior fates. We further test the hypothesis by investigating the role of cWnt signaling during early patterning along the A/P axis of the hemichordate Saccoglossus kowalevskii. We have cloned and investigated the expression of the complete Wnt ligand and Frizzled receptor complement of S. kowalevskii during early development along with many secreted Wnt modifiers. Eleven of the 13 Wnt ligands are ectodermally expressed in overlapping domains, predominantly in the posterior, and Wnt antagonists are localized predominantly to the anterior ectoderm in a pattern reminiscent of their distribution in vertebrate embryos. Overexpression and knockdown experiments, in combination with embryological manipulations, establish the importance of cWnt signaling for repression of anterior fates and activation of mid-axial ectodermal fates during the early development of S. kowalevskii. However, surprisingly, terminal posterior fates, defined by posterior Hox genes, are unresponsive to manipulation of cWnt levels during the early establishment of the A/P axis at late blastula and early gastrula. We establish experimental support for a conserved role of Wnt signaling in the early specification of the A/P axis during deuterostome body plan diversification, and further build support for an ancestral role of this pathway in early evolution of the bilaterian A/P axis. We find strong support for a role of cWnt in suppression of anterior fates and promotion of mid-axial fates, but we find no evidence that cWnt signaling plays a role in the early specification of the most posterior axial fates in S. kowalevskii. This posterior autonomy may be a conserved feature of early deuterostome axis specification.


Assuntos
Linhagem da Célula/fisiologia , Desenvolvimento Embrionário/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Transporte Biológico , Padronização Corporal/fisiologia , Ectoderma , Receptores Frizzled/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genes Homeobox , Homeostase , Planárias , Poliquetos/embriologia , Poliquetos/fisiologia
9.
J Cell Sci ; 130(7): 1333-1340, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28202692

RESUMO

Gap junction turnover occurs through the internalization of both of the plasma membranes of a gap junction plaque, forming a double membrane-enclosed vesicle, or connexosome. Phosphorylation has a key role in regulation, but further progress requires the ability to clearly distinguish gap junctions and connexosomes, and to precisely identify proteins associated with them. We examined, by using electron microscopy, serial sections of mouse preovulatory ovarian follicles that had been collected with an automated tape collecting ultramicrotome (ATUM). We found that connexosomes can form from adjacent cell bodies, from thin cell processes or from the same cell. By immunolabeling serial sections, we found that residue S368 of connexin 43 (also known as GJA1) is phosphorylated on gap junctions and connexosomes, whereas connexin 43 residue S262 is phosphorylated only on some connexosomes. These data suggest that phosphorylation at S262 contributes to connexosome formation or processing, and they provide more precise evidence that phosphorylation has a key role in gap junction internalization. Serial section electron microscopy of immunogold-labeled tissues offers a new way to investigate the three-dimensional organization of cells in their native environment.


Assuntos
Conexina 43/metabolismo , Microscopia Eletrônica/métodos , Animais , Feminino , Junções Comunicantes/metabolismo , Junções Comunicantes/ultraestrutura , Cavalos , Camundongos Endogâmicos C57BL , Fosforilação , Fosfosserina/metabolismo , Coloração e Rotulagem
10.
Proc Natl Acad Sci U S A ; 112(17): 5527-32, 2015 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-25775542

RESUMO

Meiosis in mammalian oocytes is paused until luteinizing hormone (LH) activates receptors in the mural granulosa cells of the ovarian follicle. Prior work has established the central role of cyclic GMP (cGMP) from the granulosa cells in maintaining meiotic arrest, but it is not clear how binding of LH to receptors that are located up to 10 cell layers away from the oocyte lowers oocyte cGMP and restarts meiosis. Here, by visualizing intercellular trafficking of cGMP in real-time in live follicles from mice expressing a FRET sensor, we show that diffusion of cGMP through gap junctions is responsible not only for maintaining meiotic arrest, but also for rapid transmission of the signal that reinitiates meiosis from the follicle surface to the oocyte. Before LH exposure, the cGMP concentration throughout the follicle is at a uniformly high level of ∼2-4 µM. Then, within 1 min of LH application, cGMP begins to decrease in the peripheral granulosa cells. As a consequence, cGMP from the oocyte diffuses into the sink provided by the large granulosa cell volume, such that by 20 min the cGMP concentration in the follicle is uniformly low, ∼100 nM. The decrease in cGMP in the oocyte relieves the inhibition of the meiotic cell cycle. This direct demonstration that a physiological signal initiated by a stimulus in one region of an intact tissue can travel across many layers of cells via cyclic nucleotide diffusion through gap junctions could provide a general mechanism for diverse cellular processes.


Assuntos
GMP Cíclico/metabolismo , Junções Comunicantes/metabolismo , Células da Granulosa/metabolismo , Meiose/fisiologia , Oócitos/metabolismo , Animais , GMP Cíclico/genética , Feminino , Junções Comunicantes/genética , Células da Granulosa/citologia , Hormônio Luteinizante/farmacologia , Meiose/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Oócitos/citologia
11.
Arch Oral Biol ; 60(5): 715-23, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25748393

RESUMO

OBJECTIVE: Signalling via ß-adrenergic receptors activates heterotrimeric G-proteins, which dissociate into α and ßγ subunits. In salivary glands, the α subunit of Gs stimulates adenylate cyclase, increasing cyclic AMP levels and promoting exocytosis. The goals of this study were to determine Gαs localization in salivary glands and whether it undergoes redistribution upon activation. METHODS: Mouse parotid and submandibular (SMG) glands were fixed with paraformaldehyde and prepared for immunofluorescence labelling with anti-Gαs. RESULTS: In unstimulated parotid and SMG acinar cells, Gαs was localized mainly to basolateral membranes. Some parotid acinar cells also exhibited cytoplasmic fluorescence. Isoproterenol (IPR) stimulation resulted in decreased membrane fluorescence and increased cytoplasmic fluorescence, which appeared relatively uniform by 30 min. Beginning about 2 h after IPR, cytoplasmic fluorescence decreased and membrane fluorescence increased, approaching unstimulated levels in SMG acini by 4 h. Some parotid acini exhibited cytoplasmic fluorescence up to 8 h after IPR. The IPR-induced redistribution of Gαs was prevented (SMG) or reduced (parotid) by prior injection of propranolol. Striated duct cells of unstimulated mice exhibited general cytoplasmic fluorescence, which was unchanged after IPR. CONCLUSIONS: Gαs is localized to basolateral membranes of unstimulated salivary acinar cells. Activation of Gαs causes its release from the cell membrane and movement into the cytoplasm. Reassociation of Gαs with the membrane begins about 2 h after stimulation in the SMG, but complete reassociation takes several hours in the parotid gland. The presence of Gαs in striated duct cells suggests a role in signal transduction of secretion and/or electrolyte transport processes.


Assuntos
Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Isoproterenol/farmacologia , Glândula Parótida/metabolismo , Glândula Submandibular/metabolismo , Células Acinares/efeitos dos fármacos , Células Acinares/metabolismo , Adenilil Ciclases/metabolismo , Animais , AMP Cíclico/metabolismo , Camundongos , Microscopia de Fluorescência , Glândula Parótida/efeitos dos fármacos , Propranolol/farmacologia , Transdução de Sinais/efeitos dos fármacos , Glândula Submandibular/efeitos dos fármacos
12.
Development ; 140(5): 1024-33, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23344709

RESUMO

FGFs act in vertebrate mesoderm induction and also play key roles in early mesoderm formation in ascidians and amphioxus. However, in sea urchins initial characterizations of FGF function do not support a role in early mesoderm induction, making the ancestral roles of FGF signaling and mechanisms of mesoderm specification in deuterostomes unclear. In order to better characterize the evolution of mesoderm formation, we have examined the role of FGF signaling during mesoderm development in Saccoglossus kowalevskii, an experimentally tractable representative of hemichordates. We report the expression of an FGF ligand, fgf8/17/18, in ectoderm overlying sites of mesoderm specification within the archenteron endomesoderm. Embryological experiments demonstrate that mesoderm induction in the archenteron requires contact with ectoderm, and loss-of-function experiments indicate that both FGF ligand and receptor are necessary for mesoderm specification. fgf8/17/18 gain-of-function experiments establish that FGF8/17/18 is sufficient to induce mesoderm in adjacent endomesoderm. These experiments suggest that FGF signaling is necessary from the earliest stages of mesoderm specification and is required for all mesoderm development. Furthermore, they suggest that the archenteron is competent to form mesoderm or endoderm, and that FGF signaling from the ectoderm defines the location and amount of mesoderm. When considered in a comparative context, these data support a phylogenetically broad requirement for FGF8/17/18 signaling in mesoderm specification and suggest that FGF signaling played an ancestral role in deuterostome mesoderm formation.


Assuntos
Cordados/embriologia , Fatores de Crescimento de Fibroblastos/fisiologia , Mesoderma/embriologia , Animais , Cordados/genética , Cordados/metabolismo , Ectoderma/embriologia , Ectoderma/metabolismo , Embrião não Mamífero , Indução Embrionária/genética , Indução Embrionária/fisiologia , Endoderma/embriologia , Endoderma/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Gastrulação/genética , Gastrulação/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/metabolismo , Modelos Biológicos , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
13.
J Membr Biol ; 245(5-6): 291-301, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22729691

RESUMO

Connexin43 (Cx43) forms gap junctions that couple the granulosa cells of ovarian follicles. In Cx43 knockout mice, follicle growth is restricted as a result of impaired granulosa cell proliferation. We have used these mice to examine the importance of specific Cx43 phosphorylation sites in follicle growth. Serines at residues 255, 262, 279, and 282 are MAP kinase substrates that, when phosphorylated, reduce junctional conductance. Mutant forms of Cx43 were constructed with these serines replaced with amino acids that cannot be phosphorylated. These mutants were transduced into Cx43 knockout ovarian somatic cells that were combined with wild-type oocytes and grafted into immunocompromised female mice permitting follicle growth in vivo. Despite residues 255 or 262 being mutated to prevent their being phosphorylated, recombinant ovaries constructed with these mutants were able to rescue the null phenotype, restoring complete folliculogenesis. In contrast, Cx43 with serine to alanine mutations at both residues 279 and 282 or at all four residues failed to rescue folliculogenesis; the mutant molecules were largely confined to intracellular sites, with few gap junctions. Using an in vitro proliferation assay, we confirmed a decrease in proliferation of granulosa cells expressing the double mutant construct. These results indicate that Cx43 phosphorylation by MAP kinase at serines 279 and 282 occurs in granulosa cells of early follicles and that this is involved in regulating follicle development.


Assuntos
Conexina 43/metabolismo , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Folículo Ovariano/citologia , Folículo Ovariano/metabolismo , Ovário/citologia , Ovário/metabolismo , Serina/metabolismo , Animais , Proliferação de Células , Feminino , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ovário/crescimento & desenvolvimento , Fosforilação
14.
Dev Biol ; 366(2): 308-16, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22546688

RESUMO

In preovulatory ovarian follicles of mice, meiotic prophase arrest in the oocyte is maintained by cyclic GMP from the surrounding granulosa cells that diffuses into the oocyte through gap junctions. The cGMP is synthesized in the granulosa cells by the transmembrane guanylyl cyclase natriuretic peptide receptor 2 (NPR2) in response to the agonist C-type natriuretic peptide (CNP). In response to luteinizing hormone (LH), cGMP in the granulosa cells decreases, and as a consequence, oocyte cGMP decreases and meiosis resumes. Here we report that within 20 min, LH treatment results in decreased guanylyl cyclase activity of NPR2, as determined in the presence of a maximally activating concentration of CNP. This occurs by a process that does not reduce the amount of NPR2 protein. We also show that by a slower process, first detected at 2h, LH decreases the amount of CNP available to bind to the receptor. Both of these LH actions contribute to decreasing cGMP in the follicle, thus signaling meiotic resumption in the oocyte.


Assuntos
Hormônio Luteinizante/metabolismo , Oócitos/metabolismo , Folículo Ovariano/enzimologia , Receptores do Fator Natriurético Atrial/metabolismo , Animais , GMP Cíclico/metabolismo , Feminino , Células da Granulosa/metabolismo , Hormônio Luteinizante/farmacologia , Meiose/efeitos dos fármacos , Camundongos , Peptídeo Natriurético Tipo C/metabolismo , Peptídeo Natriurético Tipo C/farmacologia , Oócitos/citologia , Folículo Ovariano/citologia , Receptores do Fator Natriurético Atrial/antagonistas & inibidores
15.
Biochim Biophys Acta ; 1818(8): 1985-92, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21819962

RESUMO

Gap junctions, composed of proteins from the connexin family, allow for intercellular communication between cells in essentially all tissues. There are 21 connexin genes in the human genome and different tissues express different connexin genes. Most connexins are known to be phosphoproteins. Phosphorylation can regulate connexin assembly into gap junctions, gap junction turnover and channel gating. Given the importance of gap junctions in development, proliferation and carcinogenesis, regulation of gap junction phosphorylation in response to wounding, hypoxia and other tissue insults is proving to be critical for cellular response and return to homeostasis. Connexin43 (Cx43) is the most widely and highly expressed gap junction protein, both in cell culture models and in humans, thus more research has been done on it and more reagents to it are available. In particular, antibodies that can report Cx43 phosphorylation status have been created allowing temporal examination of specific phosphorylation events in vivo. This review is focused on the use of these antibodies in tissue in situ, predominantly looking at Cx43 phosphorylation in brain, heart, endothelium and epithelium with reference to other connexins where data is available. These data allow us to begin to correlate specific phosphorylation events with changes in cell and tissue function. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and characteristics.


Assuntos
Encéfalo/metabolismo , Conexina 43/fisiologia , Endotélio/metabolismo , Epitélio/metabolismo , Junções Comunicantes/fisiologia , Miocárdio/metabolismo , Animais , Conexina 43/metabolismo , Genoma Humano , Homeostase , Humanos , Modelos Biológicos , Neoplasias/metabolismo , Fosforilação , Transdução de Sinais , Distribuição Tecidual
16.
Reproduction ; 140(5): 655-62, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20826538

RESUMO

The meiotic cell cycle in mouse oocytes is arrested in prophase, and then restarted when LH acts on the surrounding granulosa cells. The granulosa cells keep meiosis arrested by providing a source of cGMP that diffuses into the oocyte through gap junctions, and LH restarts the cell cycle by closing the junctions and by decreasing granulosa cell cGMP, thus lowering oocyte cGMP. Epidermal growth factor receptor (EGFR) activation is an essential step in triggering LH-induced meiotic resumption, but its relationship to the cGMP decrease in the follicle is incompletely understood, and its possible function in causing gap junction closure has not been investigated. Here, we use EGFR agonists (epiregulin and amphiregulin) and an EGFR kinase inhibitor (AG1478) to study the function of the EGFR in the signaling pathways leading to the release of oocytes from prophase arrest. Our results indicate that the EGFR kinase contributes to LH-induced meiotic resumption in two different ways. First, it is required for gap junction closure. Second, it is required for an essential component of the decrease in follicle cGMP. Our data show that the EGFR kinase-dependent component of the cGMP decrease is required for LH-induced meiotic resumption, but they also indicate that an as yet unidentified pathway accounts for a large part of the cGMP decrease.


Assuntos
GMP Cíclico/fisiologia , Receptores ErbB/fisiologia , Junções Comunicantes/fisiologia , Hormônio Luteinizante/fisiologia , Meiose/fisiologia , Folículo Ovariano/fisiologia , Anfirregulina , Animais , Família de Proteínas EGF , Fator de Crescimento Epidérmico/farmacologia , Epirregulina , Feminino , Glicoproteínas/farmacologia , Immunoblotting , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Quinazolinas , Tirfostinas/farmacologia
17.
Development ; 136(11): 1869-78, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19429786

RESUMO

Mammalian oocytes are arrested in meiotic prophase by an inhibitory signal from the surrounding somatic cells in the ovarian follicle. In response to luteinizing hormone (LH), which binds to receptors on the somatic cells, the oocyte proceeds to second metaphase, where it can be fertilized. Here we investigate how the somatic cells regulate the prophase-to-metaphase transition in the oocyte, and show that the inhibitory signal from the somatic cells is cGMP. Using FRET-based cyclic nucleotide sensors in follicle-enclosed mouse oocytes, we find that cGMP passes through gap junctions into the oocyte, where it inhibits the hydrolysis of cAMP by the phosphodiesterase PDE3A. This inhibition maintains a high concentration of cAMP and thus blocks meiotic progression. LH reverses the inhibitory signal by lowering cGMP levels in the somatic cells (from approximately 2 microM to approximately 80 nM at 1 hour after LH stimulation) and by closing gap junctions between the somatic cells. The resulting decrease in oocyte cGMP (from approximately 1 microM to approximately 40 nM) relieves the inhibition of PDE3A, increasing its activity by approximately 5-fold. This causes a decrease in oocyte cAMP (from approximately 700 nM to approximately 140 nM), leading to the resumption of meiosis.


Assuntos
AMP Cíclico/metabolismo , GMP Cíclico/fisiologia , Meiose/fisiologia , Oócitos/fisiologia , Animais , Células Cultivadas , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3 , Feminino , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/fisiologia , Humanos , Hormônio Luteinizante/farmacologia , Hormônio Luteinizante/fisiologia , Meiose/efeitos dos fármacos , Camundongos , Oócitos/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/fisiologia
18.
Methods Mol Biol ; 518: 157-73, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19085139

RESUMO

The mammalian oocyte develops within a complex of somatic cells known as a follicle, within which signals from the somatic cells regulate the oocyte, and signals from the oocyte regulate the somatic cells. Because isolation of the oocyte from the follicle disrupts these communication pathways, oocyte physiology is best studied within an intact follicle. Here we describe methods for quantitative microinjection of follicle-enclosed mouse oocytes, thus allowing the introduction of signaling molecules as well as optical probes into the oocyte within its physiological environment.


Assuntos
Microinjeções/métodos , Oócitos/metabolismo , Folículo Ovariano/citologia , Animais , Dióxido de Carbono , Células Cultivadas , Dissecação , Feminino , Umidade , Camundongos , Oócitos/citologia , Folículo Ovariano/metabolismo , Temperatura
19.
Development ; 135(19): 3229-38, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18776144

RESUMO

Luteinizing hormone (LH) acts on ovarian follicles to reinitiate meiosis in prophase-arrested mammalian oocytes, and this has been proposed to occur by interruption of a meioisis-inhibitory signal that is transmitted through gap junctions into the oocyte from the somatic cells that surround it. To investigate this idea, we microinjected fluorescent tracers into live antral follicle-enclosed mouse oocytes, and we demonstrate for the first time that LH causes a decrease in the gap junction permeability between the somatic cells, prior to nuclear envelope breakdown (NEBD). The decreased permeability results from the MAP kinase-dependent phosphorylation of connexin 43 on serines 255, 262 and 279/282. We then tested whether the inhibition of gap junction communication was sufficient and necessary for the reinitiation of meiosis. Inhibitors that reduced gap junction permeability caused NEBD, but an inhibitor of MAP kinase activation that blocked gap junction closure in response to LH did not prevent NEBD. Thus, both MAP kinase-dependent gap junction closure and another redundant pathway function in parallel to ensure that meiosis resumes in response to LH.


Assuntos
Conexina 43/metabolismo , Junções Comunicantes/efeitos dos fármacos , Hormônio Luteinizante/farmacologia , Meiose/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/metabolismo , Animais , Permeabilidade da Membrana Celular/efeitos dos fármacos , Conexina 43/química , Células do Cúmulo/citologia , Células do Cúmulo/efeitos dos fármacos , Células do Cúmulo/metabolismo , Feminino , Junções Comunicantes/metabolismo , Células da Granulosa/citologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Meiose/fisiologia , Camundongos , Oócitos/citologia , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Folículo Ovariano/citologia , Fosforilação , Serina/química , Técnicas de Cultura de Tecidos
20.
Dev Biol ; 310(2): 240-9, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17850783

RESUMO

The maintenance of meiotic prophase arrest in fully grown vertebrate oocytes depends on the activity of a G(s) G-protein that activates adenylyl cyclase and elevates cAMP, and in the mouse oocyte, G(s) is activated by a constitutively active orphan receptor, GPR3. To determine whether the action of luteinizing hormone (LH) on the mouse ovarian follicle causes meiotic resumption by inhibiting GPR3-G(s) signaling, we examined the effect of LH on the localization of Galpha(s). G(s) activation in response to stimulation of an exogenously expressed beta(2)-adrenergic receptor causes Galpha(s) to move from the oocyte plasma membrane into the cytoplasm, whereas G(s) inactivation in response to inhibition of the beta(2)-adrenergic receptor causes Galpha(s) to move back to the plasma membrane. However, LH does not cause a change in Galpha(s) localization, indicating that LH does not act by terminating receptor-G(s) signaling.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Hormônio Luteinizante/farmacologia , Meiose/fisiologia , Oócitos/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Membrana Celular/metabolismo , Células Cultivadas , Cruzamentos Genéticos , Feminino , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Oócitos/efeitos dos fármacos , Transporte Proteico , Receptores Adrenérgicos beta 2/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA