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











Base de dados
Intervalo de ano de publicação
1.
Reprod Fertil Dev ; 29(3): 609-620, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26488911

RESUMO

A dynamic partnership between follicle-stimulating hormone (FSH) and activin is required for normal Sertoli cell development and fertility. Disruptions to this partnership trigger Sertoli cells to deviate from their normal developmental pathway, as observed in inhibin α-knockout (Inha-KO) mice, which feature Sertoli cell tumours in adulthood. Here, we identified the developmental windows by which adult Sertoli cell tumourigenesis is most FSH sensitive. FSH was suppressed for 7 days in Inha-KO mice and wild-type littermates during the 1st, 2nd or 4th week after birth and culled in the 5th week to assess the effect on adult Sertoli cell development. Tumour growth was profoundly reduced in adult Inha-KO mice in response to FSH suppression during Weeks 1 and 2, but not Week 4. Proliferative Sertoli cells were markedly reduced in adult Inha-KO mice following FSH suppression during Weeks 1, 2 or 4, resulting in levels similar to those in wild-type mice, with greatest effect observed at the 2 week time point. Apoptotic Sertoli cells increased in adult Inha-KO mice after FSH suppression during Week 4. In conclusion, acute FSH suppression during the 1st or 2nd week after birth in Inha-KO mice profoundly suppresses Sertoli cell tumour progression, probably by inhibiting proliferation in the adult, with early postnatal Sertoli cells being most sensitive to FSH action.


Assuntos
Inibinas/metabolismo , Tumor de Células de Sertoli/patologia , Espermatogênese/genética , Neoplasias Testiculares/patologia , Ativinas/sangue , Animais , Hormônio Foliculoestimulante/sangue , Inibinas/genética , Masculino , Camundongos , Camundongos Knockout , Tumor de Células de Sertoli/genética , Tumor de Células de Sertoli/metabolismo , Células de Sertoli/metabolismo , Células de Sertoli/patologia , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo , Testículo/metabolismo , Testículo/patologia
2.
Mol Cell Endocrinol ; 422: 84-92, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26687063

RESUMO

Mature TGF-ß proteins are used in vivo to promote bone growth, combat obesity, reverse fibrosis and pulmonary arterial hypertension, and as potential rejuvenation factors. However, the serum half-life of this family of growth factors is short (∼5 min), limiting their therapeutic potential. Because TGF-ß proteins are normally secreted from cells with their prodomains attached, we considered whether these molecules could extend the in vivo half-life and activity of their respective growth factors. Using activin A as a model ligand, we initially modified the cleavage site between the pro- and mature domains to ensure complete processing of the activin A precursor. Co-immunoprecipitation studies confirmed mature activin A is secreted from cells in a non-covalent complex with its prodomain, however, the affinity of this interaction is not sufficient to suppress activin A in vitro biological activity. The plasma clearance profiles of purified pro- and mature activin A were determined over a 4 h period in adult male rats. Both activin forms demonstrated a two-phase decay, with the half-life of pro-activin A (t1/2 fast = 12.5 min, slow = 31.0 min) being greater than that of mature activin A (t1/2 fast = 5.5 min, slow = 20.3 min). Both pro- and mature activin A induced significant increases in serum follicle stimulating hormone levels after 4 h, but no differences were observed in the relative in vivo bioactivities of the two activin isoforms. Increased serum half-life of activin A in the presence of its prodomain identifies a new means to increase the therapeutic effectiveness of TGF-ß proteins.


Assuntos
Subunidades beta de Inibinas/química , Subunidades beta de Inibinas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Hormônio Foliculoestimulante/sangue , Meia-Vida , Subunidades beta de Inibinas/farmacologia , Masculino , Modelos Moleculares , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacologia , Estrutura Terciária de Proteína , Ratos
3.
J Med Chem ; 58(14): 5637-48, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-26098096

RESUMO

Activin belongs to the TGFß superfamily, which is associated with several disease conditions, including cancer-related cachexia, preterm labor with delivery, and osteoporosis. Targeting activin and its related signaling pathways holds promise as a therapeutic approach to these diseases. A small-molecule ligand-binding groove was identified in the interface between the two activin ßA subunits and was used for a virtual high-throughput in silico screening of the ZINC database to identify hits. Thirty-nine compounds without significant toxicity were tested in two well-established activin assays: FSHß transcription and HepG2 cell apoptosis. This screening workflow resulted in two lead compounds: NUCC-474 and NUCC-555. These potential activin antagonists were then shown to inhibit activin A-mediated cell proliferation in ex vivo ovary cultures. In vivo testing showed that our most potent compound (NUCC-555) caused a dose-dependent decrease in FSH levels in ovariectomized mice. The Blitz competition binding assay confirmed target binding of NUCC-555 to the activin A:ActRII that disrupts the activin A:ActRII complex's binding with ALK4-ECD-Fc in a dose-dependent manner. The NUCC-555 also specifically binds to activin A compared with other TGFß superfamily member myostatin (GDF8). These data demonstrate a new in silico-based strategy for identifying small-molecule activin antagonists. Our approach is the first to identify a first-in-class small-molecule antagonist of activin binding to ALK4, which opens a completely new approach to inhibiting the activity of TGFß receptor superfamily members. in addition, the lead compound can serve as a starting point for lead optimization toward the goal of a compound that may be effective in activin-mediated diseases.


Assuntos
Ativinas/antagonistas & inibidores , Ensaios de Triagem em Larga Escala , Interface Usuário-Computador , Ativinas/química , Ativinas/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cristalografia por Raios X , Avaliação Pré-Clínica de Medicamentos , Feminino , Hormônio Foliculoestimulante/antagonistas & inibidores , Células Hep G2 , Humanos , Camundongos , Simulação de Acoplamento Molecular , Ovário/citologia , Ovário/efeitos dos fármacos , Conformação Proteica , Transdução de Sinais/efeitos dos fármacos
4.
Am J Physiol Endocrinol Metab ; 306(8): E893-903, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24569591

RESUMO

Oxygen tension is critical for follicle growth and metabolism, especially for early-stage follicles, where vascularity is limited. Its role and underlying mechanism in the in vitro activation and maturation of immature to ovulatory follicles is largely unknown. In this study, early secondary (110 µm) murine follicles were isolated and encapsulated in alginate hydrogels to replicate the in vivo environment of the growing/maturing follicle. Encapsulated follicles were cultured for 8 days at either 2.5 or 20% O2. Survival (2.6-fold) and growth (1.2-fold) were significantly higher for follicles cultured at 2.5% compared with 20% O2. Using a mouse hypoxia-signaling pathway qRT-PCR array and GeneGo Metacore analysis, we found that direct target genes of the hypoxia-activated HIF1-complex were significantly upregulated in follicles cultured for 8 days at 2.5% compared with 20% O2, including the carbohydrate transport and metabolism genes Slc2a3, Vegfa, Slc2a1, Edn1, Pgk1, Ldha, and Hmox1. Other upregulated genes included carbohydrate transporters (Slc2a1, Slc2a3, and Slc16a3) and enzymes essential for glycolysis (Pgk1, Hmox1, Hk2, Gpi1, Pfkl, Pfkp, Aldoa, Gapdh, Pgam1, Eno1, Pkm2, and Ldha). For follicles cultured at 2.5% O2, a 7.2-fold upregulation of Vegfa correlated to an 18-fold increase in VEGFA levels, and a 3.2-fold upregulation of Ldha correlated to a 4.8-fold increase in lactate levels. Both VEGFA and lactate levels were significantly higher in follicles cultured at 2.5% compared with 20% O2. Therefore, enhanced hypoxia-mediated glycolysis is essential for growth and survival of early secondary follicles and provides vital insights into improving in vitro culture conditions.


Assuntos
Metabolismo dos Carboidratos , Folículo Ovariano/crescimento & desenvolvimento , Animais , Hormônio Antimülleriano/metabolismo , Transporte Biológico/efeitos dos fármacos , Metabolismo dos Carboidratos/efeitos dos fármacos , Hipóxia Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Estradiol/metabolismo , Feminino , Inibinas/metabolismo , Camundongos , Oogênese/fisiologia , Folículo Ovariano/efeitos dos fármacos , Oxigênio/farmacologia
5.
FASEB J ; 28(4): 1711-23, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24378873

RESUMO

In models of cancer cachexia, inhibiting type IIB activin receptors (ActRIIBs) reverse muscle wasting and prolongs survival, even with continued tumor growth. ActRIIB mediates signaling of numerous TGF-ß proteins; of these, we demonstrate that activins are the most potent negative regulators of muscle mass. To determine whether activin signaling in the absence of tumor-derived factors induces cachexia, we used recombinant serotype 6 adeno-associated virus (rAAV6) vectors to increase circulating activin A levels in C57BL/6 mice. While mice injected with control vector gained ~10% of their starting body mass (3.8±0.4 g) over 10 wk, mice injected with increasing doses of rAAV6:activin A exhibited weight loss in a dose-dependent manner, to a maximum of -12.4% (-4.2±1.1 g). These reductions in body mass in rAAV6:activin-injected mice correlated inversely with elevated serum activin A levels (7- to 24-fold). Mechanistically, we show that activin A reduces muscle mass and function by stimulating the ActRIIB pathway, leading to deleterious consequences, including increased transcription of atrophy-related ubiquitin ligases, decreased Akt/mTOR-mediated protein synthesis, and a profibrotic response. Critically, we demonstrate that the muscle wasting and fibrosis that ensues in response to excessive activin levels is fully reversible. These findings highlight the therapeutic potential of targeting activins in cachexia.


Assuntos
Ativinas/genética , Caquexia/genética , Expressão Gênica , Atrofia Muscular/genética , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Ativinas/sangue , Ativinas/metabolismo , Animais , Western Blotting , Caquexia/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dependovirus/genética , Vetores Genéticos/genética , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/metabolismo , Miostatina/deficiência , Miostatina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Transdução de Sinais/genética
6.
J Pathol ; 229(4): 599-607, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23180294

RESUMO

Activins are involved in the regulation of a diverse range of physiological processes including development, reproduction, and fertility, and have been implicated in the progression of cancers. Bioactivity is regulated by the inhibin α-subunit and by an activin-binding protein, follistatin. The activin-ß(C) subunit was not considered functionally significant in this regard due to an absence of phenotype in knockout mice. However, activin-ß(C) forms heterodimers with activin-ß(A) and activin-C antagonizes activin-A in vitro. Thus, it is proposed that overexpression, rather than loss of activin-ß(C) , regulates activin-A bioactivity. In order to prove biological efficacy, inhibin α-subunit knockout mice (α-KO) were crossed with mice overexpressing activin-ß(C) (ActC++). Deletion of inhibin leads to Sertoli and granulosa cell tumours, increased activin-A, and cancer-associated cachexia. Therefore, cachexia and reproductive tumour development should be modulated in α-KO/ActC++ mice, where excessive activin-A is the underlying cause. Accordingly, a reduction in activin-A, no significant weight loss, and reduced incidence of reproductive tumours were evident in α-KO/ActC++ mice. Overexpression of activin-ß(C) antagonized the activin signalling cascade; thus, the tumourigenic effects of activin-A were abrogated. This study provides proof of the biological relevance of activin-ß(C) . Being a regulator of activin-A, it is able to abolish cachexia and modulate reproductive tumour development in α-KO mice.


Assuntos
Ativinas/metabolismo , Caquexia/genética , Subunidades beta de Inibinas/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Testiculares/genética , Ativinas/sangue , Ativinas/genética , Animais , Apoptose , Linhagem Celular , Proliferação de Células , Dimerização , Progressão da Doença , Feminino , Folistatina/metabolismo , Expressão Gênica , Humanos , Subunidades beta de Inibinas/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fosforilação , Deleção de Sequência , Proteína Smad2/metabolismo , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/patologia
7.
J Biol Chem ; 287(11): 8060-70, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22267736

RESUMO

Inhibin is a heterodimeric peptide hormone produced in the ovary that antagonizes activin signaling and FSH synthesis in the pituitary. The inhibin ß-subunit interacts with the activin type II receptor (ActRII) to functionally antagonize activin. The inhibin α-subunit mature domain (N terminus) arose relatively early during the evolution of the hormone, and inhibin function is decreased by an antibody directed against the α-subunit N-terminal extension region or by deletion of the N-terminal region. We hypothesized that the α-subunit N-terminal extension region interacts with the activin type I receptor (ALK4) to antagonize activin signaling in the pituitary. Human or chicken free α-subunit inhibited activin signaling in a pituitary gonadotrope-derived cell line (LßT2) in a dose-dependent manner, whereas an N-terminal extension deletion mutant did not. An α-subunit N-terminal peptide, but not a control peptide, was able to inhibit activin A signaling and decrease activin-stimulated FSH synthesis. Biotinylated inhibin A, but not activin A, bound ALK4. Soluble ALK4-ECD bioneutralized human free α-subunit in LßT2 cells, but did not affect activin A function. Competitive binding ELISAs with N-terminal mutants and an N-terminal region peptide confirmed that this region is critical for direct interaction of the α-subunit with ALK4. These data expand our understanding of how endocrine inhibin achieves potent antagonism of local, constitutive activin action in the pituitary, through a combined mechanism of competitive binding of both ActRII and ALK4 by each subunit of the inhibin heterodimer, in conjunction with the co-receptor betaglycan, to block activin receptor-ligand binding, complex assembly, and downstream signaling.


Assuntos
Receptores de Ativinas Tipo I/metabolismo , Inibinas/metabolismo , Transdução de Sinais/fisiologia , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Animais , Células CHO , Galinhas , Cricetinae , Cricetulus , Humanos , Inibinas/genética , Masculino , Camundongos , Mutação , Peptídeos/genética , Peptídeos/metabolismo
8.
Mol Cell Endocrinol ; 359(1-2): 2-12, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21763751

RESUMO

Like other members of the transforming growth factor-ß (TGF-ß) superfamily, activins are synthesised as precursor molecules comprising an N-terminal prodomain and C-terminal mature region. During synthesis, the prodomain interacts non-covalently with mature activin, maintaining the molecule in a conformation competent for dimerisation. Dimeric precursors are cleaved by proprotein convertases and activin is secreted from the cell non-covalently associated with its propeptide. Extracellularly, the propeptide interacts with heparan sulfate proteoglycans to regulate activin localization within tissues. The mature activin dimer exhibits the classic 'open-hand' structure of TGF-ß ligands with 'finger-like' domains projecting outward from the cysteine knot core of the molecule. These finger domains form the binding epitopes for type I and II serine/threonine kinase receptors. Activins ability to access its signalling receptors is regulated by the extracellular binding proteins, follistatin, follistatin-like-3, and by inhibins, which, in the presence of betaglycan, sequester type II receptors.


Assuntos
Ativinas/fisiologia , Precursores de Proteínas/fisiologia , Receptores de Ativinas Tipo I/metabolismo , Receptores de Ativinas Tipo I/fisiologia , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Receptores de Activinas Tipo II/fisiologia , Ativinas/química , Ativinas/metabolismo , Motivos de Aminoácidos , Animais , Humanos , Inibinas/metabolismo , Inibinas/fisiologia , Ligação Proteica , Precursores de Proteínas/metabolismo , Estrutura Terciária de Proteína , Proteoglicanas/metabolismo , Proteoglicanas/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Transdução de Sinais
9.
Endocrinology ; 152(10): 3758-68, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21750050

RESUMO

Elevated activin A levels in inhibin-deficient mice promote the development of gonadal tumors and induce cachexia by reducing muscle, liver, stomach, and fat mass. Because activin A is an important regulator of tissue growth, inhibiting the actions of this TGFß family ligand may halt or reverse pathology in diseased tissues. In this study, we modified the activin A propeptide to generate a specific activin antagonist. Propeptides mediate the synthesis and secretion of all TGFß ligands and, for some family members (e.g. TGFß1), bind the mature growth factor with high enough affinity to confer latency. By linking the C-terminal region of the TGFß1 propeptide to the N-terminal region of the activin A propeptide, we generated a chimeric molecule [activin/TGFß1 propeptide (AT propeptide)] with increased affinity for activin A. The AT propeptide was 30-fold more potent than the activin A propeptide at suppressing activin-induced FSH release by LßT2 pituitary gonadotrope cells. Binding of the AT propeptide to activin A shields the type II receptor binding site, thereby reducing Smad2 phosphorylation and downstream signaling. In comparison with the commonly used activin antagonists, follistatin (IC(50) 0.42 nM), soluble activin type II receptor A-Fc (IC(50) 0.47 nM), and soluble activin type II receptor B-Fc (IC(50) 0.91 nM), the AT propeptide (IC(50) 2.6 nM) was slightly less potent. However, it was more specific, inhibiting activin A and activin B (IC(50) 10.26 nM) but not the closely related ligands, myostatin and growth differentiation factor-11. As such, the AT propeptide represents the first specific activin antagonist, and it should be an effective reagent for blocking activin actions in vivo.


Assuntos
Ativinas/antagonistas & inibidores , Precursores de Proteínas/biossíntese , Proteínas Recombinantes de Fusão/biossíntese , Fator de Crescimento Transformador beta1/biossíntese , Ativinas/biossíntese , Sequência de Aminoácidos , Animais , Células HEK293 , Humanos , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Precursores de Proteínas/farmacologia , Proteínas Recombinantes de Fusão/farmacologia
10.
Vitam Horm ; 85: 299-321, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21353886

RESUMO

Inhibins A and B are gonadal factors that negatively regulate FSH synthesis by the anterior pituitary. Across the menstrual cycle, women show a strong inverse correlation between circulating FSH and inhibin B, estradiol, and anti-Mullerian hormone (AMH), but not with inhibin A. Estradiol is believed to provide a tonic inhibitory effect while the inhibitory role of AMH is unknown. In human males, inhibin B is the primary testicular factor regulating FSH with limited effects by gonadal steroids. In vitro and in vivo studies in rats indicate that inhibin B is more biologically active than inhibin A but showed a lower affinity for the activin type II receptors and the co-receptor, betaglycan, suggesting an alternative mechanism. While this review reinforces the important role inhibin plays in regulating FSH, the observed differences in mode of action of inhibins A and B and their interplay with other gonadal factors are still poorly understood.


Assuntos
Retroalimentação Fisiológica , Hormônio Foliculoestimulante/metabolismo , Inibinas/fisiologia , Hipófise/metabolismo , Animais , Feminino , Humanos , Masculino
11.
Immunol Cell Biol ; 89(8): 889-96, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21445090

RESUMO

Activin A, a member of the transforming growth factor-ß superfamily, is a critical early mediator of acute inflammation. Activin A release coincides with the release of tumour necrosis factor-α (TNF-α) in models of lipopolysaccharide (LPS)-induced inflammation. The source of circulating activin A during acute inflammation has not been identified and the potential contribution of leukocyte subsets was examined in the following study. Human leukocytes from healthy volunteers were fractionated using Ficoll gradients and cultured under serum-free conditions. Freshly isolated human neutrophils contained 20-fold more activin A than blood mononuclear cells as measured by enzyme-linked immunosorbent assay (ELISA), and both dimeric and monomeric forms of activin A were detected in these cells by western blotting. Activin A was predominantly immunolocalized in the neutrophil cytoplasm. Purified neutrophils secreted activin A in culture when stimulated by TNF-α, but were unable to respond to LPS directly. Although TNF-α stimulated activin A release from neutrophils within 1 h, activin subunit mRNA expression did not increase until 12 h of culture, and the amount of activin A released following TNF-α stimulation did not change between 1 and 12 h. Specific inhibition of the p38 MAP kinase signalling pathway blocked TNF-α-induced activin release, and the secretion of activin A was not due to TNF-α-induced neutrophil apoptosis. These data provide the first evidence that neutrophils are a significant source of mature, stored activin A. Stimulation of the release of neutrophil activin A by TNF-α may contribute to the early peak in circulating activin A levels during acute inflammation.


Assuntos
Ativinas/metabolismo , Neutrófilos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Ativinas/sangue , Apoptose , Ensaio de Imunoadsorção Enzimática , Humanos , Imidazóis/farmacologia , Inflamação/induzido quimicamente , Mediadores da Inflamação , Leucócitos Mononucleares/imunologia , Lipopolissacarídeos/imunologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neutrófilos/citologia , Neutrófilos/imunologia , Piridinas/farmacologia , RNA Mensageiro/biossíntese , Fator de Necrose Tumoral alfa/antagonistas & inibidores
12.
J Biol Chem ; 285(22): 17029-37, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20308061

RESUMO

Transforming growth factor-beta1 (TGF-beta1) is secreted as part of an inactive complex consisting of the mature dimer, the TGF-beta1 propeptide (latency-associated peptide (LAP)), and latent TGF-beta-binding proteins. Using in vitro mutagenesis, we identified the regions of LAP that govern the cooperative assembly and stability of the latent TGF-beta1 complex. Initially, hydrophobic LAP residues (Ile(53), Leu(54), Leu(57), and Leu(59)), which form a contiguous epitope on one surface of an amphipathic alpha-helix, interact with mature TGF-beta1 to form the small latent complex. TGF-beta1 binding is predicted to alter LAP conformation, exposing ionic residues (Arg(45), Arg(50), Lys(56), and Arg(58)) on the other side of the alpha-helix, which form the binding site for latent TGF-beta-binding proteins. The stability of the resultant large latent complex is dependent upon covalent dimerization of LAP, which is facilitated by key residues (Phe(198), Asp(199), Val(200), Leu(208), Phe(217), and Leu(219)) at the dimer interface. Significantly, genetic mutations in LAP (e.g. R218H) that cause the rare bone disorder Camurati-Engelmann disease disrupted dimerization and reduced the stability of the latent TGF-beta1 complex.


Assuntos
Proteínas de Ligação a TGF-beta Latente/química , Peptídeos/química , Fator de Crescimento Transformador beta1/metabolismo , Sequência de Aminoácidos , Bioensaio , Síndrome de Camurati-Engelmann/metabolismo , Meios de Cultivo Condicionados/farmacologia , Dimerização , Matriz Extracelular/metabolismo , Humanos , Dados de Sequência Molecular , Mutação , Ligação Proteica , Mapeamento de Interação de Proteínas/métodos , Estrutura Terciária de Proteína , Fator de Crescimento Transformador beta/metabolismo
13.
Reprod Biol Endocrinol ; 7: 122, 2009 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-19874624

RESUMO

BACKGROUND: The human endometrium is unique in its capacity to remodel constantly throughout adult reproductive life. Although the processes of tissue damage and breakdown in the endometrium have been well studied, little is known of how endometrial regeneration is achieved after menstruation. Nodal, a member of the transforming growth factor-beta superfamily, regulates the processes of pattern formation and differentiation that occur during early embryo development. METHODS: In this study, the expression of Nodal, Cripto (co-receptor) and Lefty A (antagonist) was examined by RT-PCR and immunohistochemistry across the menstrual cycle and in endometrial carcinomas. RESULTS: Nodal and Cripto were found to be expressed at high levels in both stromal and epithelial cells during the proliferative phase of the menstrual cycle. Although immunoreactivity for both proteins in surface and glandular epithelium was maintained at relatively steady-state levels across the cycle, their expression was significantly decreased within the stromal compartment by the mid-secretory phase. Lefty expression, as has previously been reported, was primarily restricted to glandular epithelium and surrounding stroma during the late secretory and menstrual phases. In line with recent studies that have shown that Nodal pathway activity is upregulated in many human cancers, we found that Nodal and Cripto immunoreactivity increased dramatically in the transition from histologic Grade 1 to histologic Grades 2 and 3 endometrial carcinomas. Strikingly, Lefty expression was low or absent in all cancer tissues. CONCLUSION: The expression of Nodal in normal and malignant endometrial cells that lack Lefty strongly supports an important role for this embryonic morphogen in the tissue remodelling events that occur across the menstrual cycle and in tumourogenesis.


Assuntos
Carcinoma/genética , Neoplasias do Endométrio/genética , Endométrio/metabolismo , Ciclo Menstrual/genética , Proteína Nodal/genética , Adulto , Líquidos Corporais/metabolismo , Carcinoma/metabolismo , Neoplasias do Endométrio/metabolismo , Endométrio/fisiologia , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Feminino , Proteínas Ligadas por GPI , Regulação Neoplásica da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Fatores de Determinação Direita-Esquerda/genética , Fatores de Determinação Direita-Esquerda/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ciclo Menstrual/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína Nodal/metabolismo , Proteína Nodal/fisiologia , Transdução de Sinais/genética , Útero/metabolismo
14.
J Biol Chem ; 283(24): 16743-51, 2008 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-18397882

RESUMO

Inhibins A and B negatively regulate the production and secretion of follicle-stimulating hormone from the anterior pituitary, control ovarian follicle development and steroidogenesis, and act as tumor suppressors in the gonads. Inhibins regulate these reproductive events by forming high affinity complexes with betaglycan and activin or bone morphogenetic protein type II receptors. In this study, the binding site of inhibin A for betaglycan was characterized using inhibin A mutant proteins. An epitope for high affinity betaglycan binding was detected spanning the outer convex surface of the inhibin alpha-subunit. Homology modeling indicates that key alpha-subunit residues (Tyr(50), Val(108), Thr(111), Ser(112), Phe(118), Lys(119), and Tyr(120)) form a contiguous epitope in this region of the molecule. Disruption of betaglycan binding by the simultaneous substitution of Thr(111), Ser(112), and Tyr(120) to alanine yielded an inhibin A variant that was unable to suppress activin-induced follicle-stimulating hormone release by rat pituitary cells in culture. Together these results indicate that a high affinity interaction between betaglycan and residues Val(108)-Tyr(120) of the inhibin alpha-subunit mediate inhibin A biological activity.


Assuntos
Inibinas/antagonistas & inibidores , Proteoglicanas/química , Receptores de Fatores de Crescimento Transformadores beta/química , Ativinas/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/química , Células CHO , Células COS , Chlorocebus aethiops , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA