Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
BMC Biotechnol ; 19(1): 20, 2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30925874

RESUMO

BACKGROUND: Baculovirus-mediated expression in insect cells is a powerful approach for protein production. However, many existing methods are time-consuming, offer limited options for protein tagging, and are unsuitable for secreted proteins requiring proteolytic maturation, such as TGF-ß family growth factors. RESULTS: To overcome the limitations of traditional baculovirus expression systems, we engineered "FlexiBAC". This system allows recombinant baculovirus formation inside insect cells and reduces the time between initial cloning and protein production to 13 days. FlexiBAC includes 143 shuttle vectors that append combinations of purification tags, fluorescent markers, proteolytic cleavage sites, trafficking signals, and chemical conjugation tags to the termini of the target protein. This system also overexpresses recombinant furin convertase to allow efficient proteolytic processing of secreted proteins. We demonstrate that FlexiBAC can be used to produce high levels of mature, active forms of TGF-ß family growth factors, such as Activin A, as well as other proteins that are typically difficult to reconstitute, such as proteins rich in coiled-coil, low complexity, and disordered domains. CONCLUSIONS: FlexiBAC is a protein expression system for production of both cytosolic proteins and secreted proteins that require proteolytic maturation. The design of FlexiBAC and its expansive complementary shuttle vector system reduces cloning steps and simplifies baculovirus production.


Assuntos
Baculoviridae/genética , Vetores Genéticos/genética , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes/genética , Ativinas/biossíntese , Ativinas/genética , Animais , Expressão Gênica , Proteínas Recombinantes/metabolismo , Spodoptera/genética , Transfecção/métodos , Cultura de Vírus/métodos
2.
Heart Lung Circ ; 28(11): 1697-1705, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30220481

RESUMO

BACKGROUND: Activin A secreted by epicardial adipose tissue (EAT) plays a major role in the progress of atrial fibrosis. However, the potential of Activin A in predicting the occurrence of postoperative atrial fibrillation (POAF) has yet to be elucidated. We aimed to investigate the predicable value of Activin A expression in EAT on POAF. METHODS: A total of 89 patients receiving cardiac surgery without atrial fibrillation (AF) history were enrolled in this study, including 49 patients with valvular heart disease (VHD) and 40 patients with non-valvular heart disease (NVHD). Activin A expression in EAT was determined by quantitative polymerase chain reaction (qPCR), while the thickness of EAT (EATT) was estimated by echocardiography. New onset POAF before discharge was documented. RESULTS: Eventually 32 patients (36.0%) developed POAF, including 20 patients with VHD (40.8%) and 12 patients with NVHD (30.0%). Activin A expression was higher in POAF than sinus rhythm (SR) patients, whether for VHD or NVHD group (All p<0.001). In general, Activin A expression predicted the occurrence of POAF with a sensitivity of 65.6% and specificity of 91.2% (AUC: 0.795; 95%CI: 0.693-0.897, p<0.001). Subgroup analysis showed that EATT was not significant for the VHD group in predicting POAF (p=0.07), while Activin A expression demonstrated a sensitivity of 60.0% and specificity of 89.7% (AUC: 0.745; 95%CI: 0.601-0.889, p<0.001). Multivariate regression analysis showed that Activin A expression in EAT was an independent risk factor for POAF (OR: OR=1.067, 95%CI:1.002-1.136, p=0.042). CONCLUSIONS: Activin A expression in EAT is an independent risk factor for POAF, which can be used for prediction of POAF, especially for patients with VHD.


Assuntos
Ativinas/biossíntese , Tecido Adiposo/metabolismo , Fibrilação Atrial/metabolismo , Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Pericárdio/metabolismo , Complicações Pós-Operatórias , Ativinas/genética , Tecido Adiposo/diagnóstico por imagem , Fibrilação Atrial/etiologia , Ecocardiografia , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Pericárdio/diagnóstico por imagem , Valor Preditivo dos Testes , Estudos Retrospectivos
3.
J Immunol ; 196(3): 1327-37, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26729812

RESUMO

Human CD14(++)CD16(-) and CD14(+/lo)CD16(+) monocyte subsets comprise 85 and 15% of blood monocytes, respectively, and are thought to represent distinct stages in the monocyte differentiation pathway. However, the differentiation fates of both monocyte subsets along the macrophage (Mϕ) lineage have not yet been elucidated. We have now evaluated the potential of CD14(++) CD16(-) and CD16(+) monocytes to differentiate and to be primed toward pro- or anti-inflammatory Mϕs upon culture with GM-CSF or M-CSF, respectively (subsequently referred to as GM14, M14, GM16, or M16). Whereas GM16 and GM14 were phenotypic and functionally analogous, M16 displayed a more proinflammatory profile than did M14. Transcriptomic analyses evidenced that genes associated with M-CSF-driven Mϕ differentiation (including FOLR2, IL10, IGF1, and SERPINB2) are underrepresented in M16 with respect to M14. The preferential proinflammatory skewing of M16 relative to M14 was found to be mediated by the secretion of activin A and the low levels of IL-10 produced by M16. In fact, activin A receptor blockade during the M-CSF-driven differentiation of CD16(+) monocytes, or addition of IL-10-containing M14-conditioned medium, significantly enhanced their expression of anti-inflammatory-associated molecules while impairing their acquisition of proinflammatory-related markers. Thus, we propose that M-CSF drives CD14(++)CD16- monocyte differentiation into bona fide anti-inflammatory Mϕs in a self-autonomous manner, whereas M-CSF-treated CD16(+) monocytes generate Mϕs with a skewed proinflammatory profile by virtue of their high activin A expression unless additional anti-inflammatory stimuli such as IL-10 are provided.


Assuntos
Ativinas/biossíntese , Diferenciação Celular/imunologia , Interleucina-10/biossíntese , Macrófagos/citologia , Monócitos/imunologia , Ativinas/imunologia , Western Blotting , Separação Celular , Citometria de Fluxo , Imunofluorescência , Humanos , Inflamação/imunologia , Interleucina-10/imunologia , Macrófagos/imunologia , Monócitos/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real , Receptores de IgG/imunologia
4.
Stem Cells ; 33(10): 3039-51, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26037810

RESUMO

Adipose stem/stromal cells (ASCs) after isolation produce numerous angiogenic growth factors. This justifies their use to promote angiogenesis per transplantation. In parallel, local coimplantation of ASC with endothelial cells (ECs) leading to formation of functional vessels by the donor cells suggests the existence of a mechanism responsible for fine-tuning ASC paracrine activity essential for vasculogenesis. As expected, conditioned media (CM) from ASC promoted ECs survival, proliferation, migration, and vasculogenesis. In contrast, media from EC-ASC cocultures had neutral effects upon EC responses. Media from cocultures exhibited lower levels of vascular endothelial growth factor (VEGF), hepatic growth factor, angiopoietin-1, and stromal cell-derived factor-1 compared with those in ASC CM. Activin A was induced in ASC in response to EC exposure and was responsible for overall antivasculogenic activity of EC-ASC CM. Except for VEGF, activin A diminished secretion of all tested factors by ASC. Activin A mediated induction of VEGF expression in ASC, but also upregulated expression of VEGF scavenger receptor FLT-1 in EC in EC-ASC cocultures. Blocking the FLT-1 expression in EC led to an increase in VEGF concentration in CM. In vitro pre-exposure of ASC to low number of EC before subcutaneous coimplantation with EC resulted in decrease in vessel density in the implants. In vitro tests suggested that activin A was partially responsible for this diminished ASC activity. This study shows that neovessel formation is associated with induction of activin A expression in ASC; this factor, by affecting the bioactivity of both ASC and EC, directs the crosstalk between these complementary cell types to establish stable vessels.


Assuntos
Ativinas/biossíntese , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais/citologia , Neovascularização Fisiológica/efeitos dos fármacos , Células Estromais/citologia , Ativinas/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Técnicas de Cocultura , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Células Estromais/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/biossíntese
5.
J Immunol ; 192(3): 1241-8, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24395917

RESUMO

The interaction of NK cells with dendritic cells (DCs) results in reciprocal cell activation through the interaction of membrane proteins and the release of soluble factors. In this article, we report that in NK-DC cocultures, among a set of 84 cytokines investigated, activin A was the second highest induced gene, with CXCL8 being the most upregulated one. Activin A is a member of the TGF-ß superfamily and was previously shown to possess both proinflammatory and anti-inflammatory activities. In NK-DC cocultures, the induction of activin A required cell contact and was dependent on the presence of proinflammatory cytokines (i.e., IFN-γ, TNF-α, and GM-CSF), as well as on NK cell-mediated DC killing. CD1(+) DCs were the main activin A producer cells among myeloid blood DC subsets. In NK-DC cocultures, inhibition of activin A by follistatin, a natural inhibitory protein, or by a specific blocking Ab, resulted in the upregulation of proinflammatory cytokine release (i.e., IL-6, IL-8, TNF-α) by DCs and in the increase of DC maturation. In conclusion, our study reports that activin A, produced during NK-DC interactions, represents a relevant negative feedback mechanism that might function to prevent excessive immune activation by DCs.


Assuntos
Ativinas/fisiologia , Células Dendríticas/imunologia , Células Matadoras Naturais/imunologia , Ativinas/biossíntese , Ativinas/genética , Animais , Antígenos CD/biossíntese , Antígenos CD/genética , Antígeno B7-2/biossíntese , Antígeno B7-2/genética , Técnicas de Cocultura , Citocinas/biossíntese , Citocinas/genética , Citotoxicidade Imunológica , Cães , Retroalimentação Fisiológica , Folistatina/farmacologia , Síndrome de Hermanski-Pudlak/patologia , Humanos , Imunoglobulinas/biossíntese , Imunoglobulinas/genética , Interleucina-15/farmacologia , Interleucina-8/biossíntese , Interleucina-8/genética , Células Matadoras Naturais/efeitos dos fármacos , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Monócitos/citologia , Monócitos/efeitos dos fármacos , Receptores de Células Matadoras Naturais/biossíntese , Receptores de Células Matadoras Naturais/genética , Regulação para Cima , Antígeno CD83
6.
Hum Mutat ; 35(3): 294-7, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24302632

RESUMO

To identify novel genetic bases of early-onset epithelial ovarian tumors, we used the trio exome sequencing strategy in a patient without familial history of cancer who presented metastatic serous ovarian adenocarcinomas at 21 years of age. We identified a single de novo mutation (c.1157A>G/p.Asn386Ser) within the INHBA gene encoding the ßA-subunit of inhibins/activins, which play a key role in ovarian development. In vitro, this mutation alters the ratio of secreted activins and inhibins. In a second patient with early-onset serous borderline papillary cystadenoma, we identified an unreported germline mutation (c.179G>T/p.Arg60Leu) of the INHA gene encoding the α-subunit, the partner of the ßA-subunit. This mutation also alters the secreted activin/inhibin ratio, by disrupting both inhibin A and inhibin B biosynthesis. In a cohort of 62 cases, we detected an additional unreported germline mutation of the INHBA gene (c.839G>A/p.Gly280Glu). Our results strongly suggest that inhibin mutations contribute to the genetic determinism of epithelial ovarian tumors.


Assuntos
Mutação em Linhagem Germinativa , Subunidades beta de Inibinas/genética , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Ovarianas/genética , Ativinas/biossíntese , Carcinoma Epitelial do Ovário , Diferenciação Celular , Estudos de Coortes , Células Epiteliais/metabolismo , Exoma , Feminino , Células da Granulosa/metabolismo , Humanos , Inibinas/biossíntese , Análise de Sequência de DNA , Adulto Jovem
7.
Am J Physiol Cell Physiol ; 306(1): C37-44, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24088895

RESUMO

Chronic myeloid leukemia (CML) is a hematopoietic stem/progenitor cell disorder in which Bcr-Abl oncoprotein inhibits cell differentiation. Differentiation induction is considered an alternative strategy for treating CML. Activin A, a member of the transforming growth factor-ß superfamily, induces erythroid differentiation of CML cells through the p38 MAPK pathway. In this study, treatment of the K562 CML stem/progenitor cell line with activin A followed by a subtoxic concentration of the Bcr-Abl inhibitor imatinib strongly induced growth inhibition and apoptosis compared with simultaneous treatment with activin A and imatinib. Imatinib-induced growth inhibition and apoptosis following activin A pretreatment were dose- and time-dependent. Imatinib-induced growth inhibition and apoptosis were also dependent on the pretreatment dose of activin A. More than 90% of the activin A-induced increases in glycophorin A-positive cells were sensitive to imatinib. However, only some of original glycophorin A-positive cells in the activin A treatment group were sensitive to imatinib. Sequential treatment with activin A and imatinib decreased Bcr-Abl, procaspase-3, Mcl-1, and Bcl-xL and also induced cleavage of procaspase-3/poly(ADP-ribose)polymerase. The reduction of erythroid differentiation in p38 MAPK dominant-negative mutants or by short hairpin RNA knockdown of p38 MAPK decreased the growth inhibition and apoptosis mediated by sequential treatment with activin A and imatinib. Furthermore, the same inhibition level of multidrug resistance 1 expression was observed in cells treated with activin A alone, treated sequentially with activin A and imatinib, or treated simultaneously with activin A and imatinib. The p38 MAPK inhibitor SB-203580 can restore activin A-inhibited multidrug resistance 1 expression. Taken together, our results suggest that a subtoxic concentration of imatinib could exhibit strong cytotoxicity against erythroid-differentiated K562 CML cells.


Assuntos
Ativinas/biossíntese , Ativinas/toxicidade , Antineoplásicos/toxicidade , Benzamidas/toxicidade , Diferenciação Celular/efeitos dos fármacos , Leucemia Mielogênica Crônica BCR-ABL Positiva , Piperazinas/toxicidade , Pirimidinas/toxicidade , Diferenciação Celular/fisiologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Mesilato de Imatinib , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Proteínas Recombinantes/toxicidade
8.
Blood ; 120(2): 431-9, 2012 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-22611157

RESUMO

Anemia is very common in patients suffering from infections or chronic inflammation and can add substantially to the morbidity of the underlying disease. It is mediated by excessive production of the iron-regulatory peptide hepcidin, but the signaling pathway responsible for hepcidin up-regulation in the inflammatory context is still not understood completely. In the present study, we show that activin B has an unexpected but crucial role in the induction of hepcidin by inflammation. There is a dramatic induction of Inhbb mRNA, encoding the activin ß(B)-subunit, in the livers of mice challenged with lipopolysaccharide, slightly preceding an increase in Smad1/5/8 phosphorylation and Hamp mRNA. Activin B also induces Smad1/5/8 phosphorylation in human hepatoma-derived cells and, synergistically with IL-6 and STAT-3 signaling, up-regulates hepcidin expression markedly, an observation confirmed in mouse primary hepatocytes. Pretreatment with a bone morphogenic protein type I receptor inhibitor showed that the effect of activin B on hepcidin expression is entirely attributable to its effect on bone morphogenetic protein signaling, most likely via activin receptor-like kinase 3. Activin B is therefore a novel and specific target for the treatment of anemia of inflammation.


Assuntos
Ativinas/biossíntese , Peptídeos Catiônicos Antimicrobianos/genética , Inflamação/metabolismo , Proteínas Smad/metabolismo , Ativinas/genética , Animais , Proteína Morfogenética Óssea 6/deficiência , Proteína Morfogenética Óssea 6/genética , Proteína Morfogenética Óssea 6/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepcidinas , Humanos , Inflamação/etiologia , Interleucina-6/deficiência , Interleucina-6/genética , Interleucina-6/metabolismo , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Regulação para Cima/efeitos dos fármacos
9.
Birth Defects Res B Dev Reprod Toxicol ; 101(5): 364-70, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25231192

RESUMO

BACKGROUND: Diabetes mellitus in pregnancy causes defects in infant heart, including the outflow tracts (OFTs). Development of the aorta and pulmonary artery, which are derived from the common OFT in the embryo, is regulated by the transforming growth factor ß (TGFß) and Wnt families, and can be perturbed by hyperglycemia-generated intracellular stress conditions. However, the underlying cellular and molecular mechanisms remain to be delineated. METHODS: Female mice were induced diabetic with streptozotocin. Embryonic and fetal OFTs were examined morphologically and histologically. Cell proliferation was assessed using 5'-bromo-2'-deoxyuridine incorporation assay. Oxidative and endoplasmic reticulum (ER) stress markers and TGFß factors were detected using immunohistochemistry. The expression of genes in the Wnt-signaling system was assessed using real-time reverse transcription polymerase chain reaction array. The role of activin-A in cell proliferation was addressed by treating embryos cultured in high glucose with activin-A. RESULTS: Maternal diabetes caused complex abnormalities in the OFTs, including aortic and pulmonary stenosis and persistent truncus arteriosus. The development of the endocardial cushions was suppressed, manifested with insufficient cellularization of the tissues. Cell proliferation was significantly decreased under oxidative and ER stress conditions. The expression of genes in the Wnt signaling was significantly altered. Activin-A and Smad3 were found to be expressed in the OFT. Treatment with activin-A rescued cell proliferation in the endocardial cushions. CONCLUSIONS: Maternal diabetes generates oxidative and ER stress conditions, suppresses TGFß and Wnt signaling, inhibits cell proliferation and cellularization of the endocardial cushions, leading to OFT septal defects. Activin-A plays a role in hyperglycemia-suppressed proliferation of the endocardial cells.


Assuntos
Diabetes Mellitus Experimental/patologia , Diabetes Gestacional/patologia , Cardiopatias Congênitas/patologia , Fator de Crescimento Transformador beta/metabolismo , Proteínas Wnt/metabolismo , Ativinas/biossíntese , Ativinas/farmacologia , Animais , Aorta/embriologia , Estenose da Valva Aórtica/patologia , Débito Cardíaco/fisiologia , Proliferação de Células , Diabetes Mellitus Experimental/induzido quimicamente , Técnicas de Cultura Embrionária , Embrião de Mamíferos/metabolismo , Coxins Endocárdicos/embriologia , Estresse do Retículo Endoplasmático , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Glucose/farmacologia , Hiperglicemia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Crista Neural/embriologia , Estresse Oxidativo , Gravidez , Artéria Pulmonar/embriologia , Estenose da Valva Pulmonar/patologia , Proteína Smad3/biossíntese , Estreptozocina , Persistência do Tronco Arterial/patologia , Via de Sinalização Wnt/genética
10.
Ontogenez ; 45(4): 272-9, 2014.
Artigo em Russo | MEDLINE | ID: mdl-25735149

RESUMO

TGFP3 family factors play an important role in regulating the balance of self-renewal and differentiation of mouse and human pluripotent stem and embryonic teratocarcinoma cells. The expression patterns of TGFbeta family signaling ligands and functional roles of these signaling pathways differ significantly in mouse and human embryonic stem cells, but the activity and functional role of these factors in mouse and human embryonic teratocarcinoma cells were not sufficiently investigated. Comparative quantitative real-time PCR analysis of the expression of TGF@[beta] family factors in mouse embryonic stem, embryonic germ, and embryonic teratocarcinoma cells showed that embryonic teratocarcinoma cells express lower ActivinA than pluripotent stem cells but similar levels of factors Nodal, Lefty 1, TGFbeta1, BMP4, and GDF3. In human nullipotent embryonic teratocarcinoma PA-1 cells, most factors of the TGFbeta family (ACTIVINA, NODAL, LEFTY 1, BMP4, and GDF3) are expressed at lower levels than in human embryonic stem cells: Thus, in mouse and human nullipotent teratocarcinoma cells, theexpression of ActivinA is significantly reduced com- pared ivith embryonic stem cells. Presumably, these differences may be associated with changes in the functional activity of the respective signaling pathways and deregulation of proliferative and antiproliferative mechanisms in embryonic teratocarcinoma cells.


Assuntos
Ativinas/biossíntese , Células-Tronco de Carcinoma Embrionário/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Teratocarcinoma/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Células-Tronco de Carcinoma Embrionário/patologia , Humanos , Camundongos , Transdução de Sinais , Teratocarcinoma/patologia
11.
J Cell Physiol ; 228(11): 2167-74, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23589129

RESUMO

In healthy bones, mineralization has to be tightly controlled to avoid pathological phenotypes. In this study, we investigated interactions between 1α,25(OH)2 D3 (1,25D3) and activin A in the regulation of osteoblast induced mineralization. In human osteoblast cultures, we demonstrated that besides stimulation of mineralization, 1,25D3 also induced activin A, a strong inhibitor of mineralization. Simultaneously, follistatin (FST), the natural antagonist of activin A, was down-regulated by1,25D3. This resulted in an increase in activin A activity during 1,25D3 treatment. We also showed that in 1,25D3-treated osteoblasts, mineralization can be further increased when activin A activity was abrogated by adding exogenous FST. This observation implies that, besides stimulation of mineralization, 1,25D3 also controls activin A-mediated inhibition of mineralization. Besides activin A, 1,25D3 also induces osteocalcin (BGLAP), another inhibitor of mineralization. Warfarin, which has been shown to inactivate osteocalcin, increased 1,25D3-induced mineralization. Interaction between these two systems became evident from the synergistic increase in BGLAP expression upon blocking activin activity in 1,25D3-treated cultures. In conclusion, we demonstrate that 1,25D3 stimulation of mineralization by human osteoblasts is suppressed by concomitant induction of inhibitors of mineralization. Mineralization induction by 1,25D3 may actually be controlled via interplay with activin A and osteocalcin. Finally, this complex regulation of mineralization substantiates the significance of tight control of mineralization to prevent excessive mineralization and consequently reduction in bone quality and strength.


Assuntos
Ativinas/biossíntese , Calcificação Fisiológica/efeitos dos fármacos , Osteoblastos/metabolismo , Vitamina D/análogos & derivados , Linhagem Celular , Folistatina/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Lectinas Tipo C/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteocalcina/genética , Osteocalcina/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Smad7/metabolismo , Vitamina D/farmacologia , Varfarina/farmacologia
12.
J Endocrinol Invest ; 36(7): 515-20, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23385491

RESUMO

BACKGROUND: Accumulating evidence supports the idea of activin A as a modulator of inflammation. In human pregnancy, elevated activin A concentrations in amniotic fluid are reported in women with intra-amniotic infection and inflammation- induced pre-term birth. AIM: To test the hypothesis that activin A was involved in the pathophysiology of amnionitis, we evaluated the effects of tumor necrosis factor-α and lipopolysaccharide on activin A production in human amniotic epithelial cells, and the effects of activin A on the expression of collagen mRNA in amniotic mesenchymal cells. MATERIALS AND METHODS: Amniotic membranes were obtained from patients without systemic disease, signs of premature delivery or fetal complications, during elective cesarean sections at term. Amniotic epithelial cells and mesenchymal cells were separately obtained by enzymatic digestion and cultured. Activin A was measured by enzyme-linked immunosorbent assay and collagen mRNA levels were assessed by quantitative PCR. RESULTS: Amniotic epithelial cells produced activin A in a cell density- and time-dependent manner. Tumor necrosis factor- α enhanced activin A production in a time-dependent (48-120 h) and dose-dependent (10-300 ng/ml) manner in amniotic epithelial cells. Lipopolysaccharide also stimulated activin A production, but the effect was less prominent. In amniotic mesenchymal cells, the effect of activin A on the expression of type I and type III collagen mRNA was suppressive. CONCLUSIONS: Tumor necrosis factor-α and lipopolysaccharide stimulated activin A production in amniotic epithelial cells, and activin A modulated expression of collagen mRNA in amniotic mesenchymal cells. These results support the idea that activin A is involved in the pathophysiology of amnionitis.


Assuntos
Ativinas/metabolismo , Âmnio/metabolismo , Colágeno Tipo III/biossíntese , Colágeno Tipo I/biossíntese , Células Epiteliais/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Ativinas/biossíntese , Âmnio/citologia , Células Cultivadas , Corioamnionite/fisiopatologia , Células Epiteliais/efeitos dos fármacos , Feminino , Humanos , Mesoderma/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
13.
Endocrinology ; 163(3)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35022746

RESUMO

Testicular-derived inhibin B (α/ß B dimers) acts in an endocrine manner to suppress pituitary production of follicle-stimulating hormone (FSH), by blocking the actions of activins (ß A/B/ß A/B dimers). Previously, we identified a homozygous genetic variant (c.1079T>C:p.Met360Thr) arising from uniparental disomy of chromosome 2 in the INHBB gene (ß B-subunit of inhibin B and activin B) in a man suffering from infertility (azoospermia). In this study, we aimed to test the causality of the p.Met360Thr variant in INHBB and testis function. Here, we used CRISPR/Cas9 technology to generate InhbbM364T/M364T mice, where mouse INHBB p.Met364 corresponds with human p.Met360. Surprisingly, we found that the testes of male InhbbM364T/M364T mutant mice were significantly larger compared with those of aged-matched wildtype littermates at 12 and 24 weeks of age. This was attributed to a significant increase in Sertoli cell and round spermatid number and, consequently, seminiferous tubule area in InhbbM364T/M364T males compared to wildtype males. Despite this testis phenotype, male InhbbM364T/M364T mutant mice retained normal fertility. Serum hormone analyses, however, indicated that the InhbbM364T variant resulted in reduced circulating levels of activin B but did not affect FSH production. We also examined the effect of this p.Met360Thr and an additional INHBB variant (c.314C>T: p.Thr105Met) found in another infertile man on inhibin B and activin B in vitro biosynthesis. We found that both INHBB variants resulted in a significant disruption to activin B in vitro biosynthesis. Together, this analysis supports that INHBB variants that limit activin B production have consequences for testis composition in males.


Assuntos
Infertilidade Masculina/genética , Subunidades beta de Inibinas/genética , Subunidades beta de Inibinas/fisiologia , Mutação , Contagem de Espermatozoides , Testículo/fisiopatologia , Ativinas/biossíntese , Ativinas/genética , Animais , Azoospermia/genética , Proteína 9 Associada à CRISPR , Hormônio Foliculoestimulante/metabolismo , Humanos , Infertilidade Masculina/fisiopatologia , Inibinas/biossíntese , Inibinas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células de Sertoli , Espermatogênese/genética , Espermatogônias , Testículo/química , Testículo/citologia
14.
J Cell Mol Med ; 15(11): 2399-410, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21143387

RESUMO

Epicardial adipose tissue (EAT) has been implicated in the development of heart disease. Nonetheless, the crosstalk between factors secreted from EAT and cardiomyocytes has not been studied. Here, we examined the effect of factors secreted from EAT on contractile function and insulin signalling in primary rat cardiomocytes. EAT and subcutaneous adipose tissue (SAT) were isolated from guinea pigs fed a high-fat (HFD) or standard diet. HFD feeding for 6 months induced glucose intolerance, and decreased fractional shortening and ejection fraction (all P < 0.05). Conditioned media (CM) generated from EAT and SAT explants were subjected to cytokine profiling using antibody arrays, or incubated with cardiomyocytes to assess the effects on insulin action and contractile function. Eleven factors were differentially secreted by EAT when compared to SAT. Furthermore, secretion of 30 factors by EAT was affected by HFD feeding. Most prominently, activin A-immunoreactivity was 6.4-fold higher in CM from HFD versus standard diet-fed animals and, 2-fold higher in EAT versus SAT. In cardiomyocytes, CM from EAT of HFD-fed animals increased SMAD2-phosphorylation, a marker for activin A-signalling, decreased sarcoplasmic-endoplasmic reticulum calcium ATPase 2a expression, and reduced insulin-mediated phosphorylation of Akt-Ser473 versus CM from SAT and standard diet-fed animals. Finally, CM from EAT of HFD-fed animals as compared to CM from the other groups markedly reduced sarcomere shortening and cytosolic Ca(2+) fluxes in cardiomyocytes. These data provide evidence for an interaction between factors secreted from EAT and cardiomyocyte function.


Assuntos
Resistência à Insulina , Miócitos Cardíacos/fisiologia , Gordura Subcutânea/fisiologia , Ativinas/biossíntese , Animais , Cálcio/análise , Doenças Cardiovasculares/complicações , Células Cultivadas , Diabetes Mellitus Tipo 2/etiologia , Gorduras na Dieta , Cobaias , Insulina/metabolismo , Miócitos Cardíacos/patologia , Pericárdio/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Endogâmicos Lew , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/biossíntese , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/fisiologia , Transdução de Sinais , Proteína Smad2/metabolismo , Gordura Subcutânea/metabolismo
15.
Dig Dis Sci ; 56(4): 1075-81, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20824496

RESUMO

BACKGROUND: Ischemia-reperfusion injury has been demonstrated in a variety of clinical settings. The morbidity associated with liver transplantation and major hepatic resections is partly a result of ischemia-reperfusion injury. Follistatin, an activin-binding protein, binds to activins and subsequently blocks their action. It was reported that blockade of the action of activin with administration of follistatin accelerates recovery from ischemia renal injury. This study was conducted to investigate the involvement of the activin-follistatin system in hepatic ischemia-reperfusion injury. METHODS: Total hepatic ischemia for 30 min was performed followed by reperfusion in a rat model. Rats were divided into two groups: a follistatin group and a control group. Follistatin (1 µg/body), which is an activin-binding protein, was administered at the time of reperfusion. RESULTS: Though 80% of animals survived in the follistatin group, four of five animals died in the control group within 3 days after reperfusion (p<0.05). AST was significantly lower at 3 h after reperfusion in the follistatin group (p<0.05). LDH was also lower at 6 h after reperfusion in the follistatin group (p<0.05). Follistatin inhibited the mRNA expression of the ßA subunit of activin. Moreover, the expression of IL-6, which is an inflammatory cytokine, was suppressed at 6 h after reperfusion in the follistatin group (p<0.05). CONCLUSIONS: The present study demonstrated that treatment with follistatin reduced the expression of IL-6 and activin resulting in beneficial support for hepatic ischemia-reperfusion injuries.


Assuntos
Folistatina/uso terapêutico , Fígado/efeitos dos fármacos , Traumatismo por Reperfusão/tratamento farmacológico , Ativinas/biossíntese , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Interleucina-6/biossíntese , L-Lactato Desidrogenase/sangue , Fígado/enzimologia , Fígado/lesões , Masculino , Ratos , Ratos Wistar , Regeneração/efeitos dos fármacos , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/mortalidade , Traumatismo por Reperfusão/patologia , Resultado do Tratamento
16.
J Neurosci Res ; 88(1): 16-23, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19681171

RESUMO

Previous studies have shown that activin A, a neuroprotective cytokine and dimeric polypeptide composed of two betaA subunits, is elevated in the cerebrospinal fluid of patients suffering from bacterial meningitis. In this study, to elucidate further the functional significance and pathophysiological implications of these findings, we demonstrated that microglial cells are not only the source but also the target cells of activin A in the central nervous system: immunohistochemistry and RT-PCR revealed expression of activin subunit betaA mRNA as well as activin receptor type I and type II mRNA in rat microglia in vitro. Further studies showed that activin enhances microglial proliferation and decreases the gamma-interferon-induced synthesis of nitric oxide, one of several microglial mediators involved in the inflammatory response in microglia activation. Furthermore, quantitative RT-PCR, Western blotting, and ELISA showed an inhibitory effect of activin on inducible nitric oxide synthase, tumor necrosis factor-alpha, interleukin-6, and interleukin-1beta gene and protein levels after lipopolysaccharide treatment. We suggest that the increased synthesis of activin A is directly involved, via influence on microglia cell functions, in the modulation of the inflammatory response in bacterial meningitis.


Assuntos
Ativinas/biossíntese , Meningites Bacterianas/metabolismo , Microglia/metabolismo , Receptores de Ativinas/genética , Receptores de Ativinas/metabolismo , Ativinas/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Animais , Western Blotting , Proliferação de Células , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imuno-Histoquímica , Interferon gama/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , Meningites Bacterianas/genética , Meningite Viral/genética , Meningite Viral/metabolismo , Microglia/efeitos dos fármacos , Pessoa de Meia-Idade , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
J Allergy Clin Immunol ; 124(3): 454-62, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19733294

RESUMO

BACKGROUND: Both transforming growth factor (TGF)-beta(1) and activin-A have been implicated in airway remodeling in asthma, but the modulation of their specific signaling pathways after disease activation remains undefined. OBJECTIVE: To define the expression kinetics of TGF-beta(1), activin-A ligands, and follistatin (a natural activin inhibitor), their type I and type II receptors (activin-like kinase[ALK]-1, ALK-5, ALK-4, TbetaRII, and ActRIIA/RIIB) and activation of signaling (via phosphorylated (p) Smad2), in the asthmatic airway after allergen challenge. METHODS: Immunohistochemistry was performed on bronchial biopsies from 15 mild atopic patients with asthma (median age, 25 years; median FEV(1)% predicted, 97%) at baseline and 24 hours after allergen inhalation. Functional effects of activin-A were evaluated by using cultured normal human bronchial epithelial (NHBE) cells. RESULTS: pSmad2(+) epithelial cells increased at 24 hours (P = .03), and pSmad2 was detected in submucosal cells. No modulation of activin-A, follistatin, or TGF-beta(1) expression was demonstrated. Activin receptor(+) cells increased after allergen challenge: ALK-4 in epithelium (P = .04) and submucosa (P = .04), and ActRIIA in epithelium (P = .01). The TGF-beta receptor ALK-5 expression was minimal in the submucosa at baseline and after challenge and was downregulated in the epithelium after challenge (P = .02), whereas ALK-1 and TbetaRII expression in the submucosa increased after allergen challenge (P = .03 and P = .004, respectively). ALK-1 and ALK-4 expression by T cells was increased after allergen challenge. Activin-A induced NHBE cell proliferation, was produced by NHBE cells in response to TNF-alpha, and downregulated TNF-alpha and IL-13-induced chemokine production by NHBE cells. CONCLUSION: Both TGF-beta and activin signaling pathways are activated on allergen provocation in asthma. Activin-A may contribute to resolution of inflammation.


Assuntos
Ativinas/biossíntese , Alérgenos/imunologia , Asma/imunologia , Testes de Provocação Brônquica , Fator de Crescimento Transformador beta/biossíntese , Receptores de Ativinas Tipo I/imunologia , Receptores de Ativinas Tipo I/metabolismo , Receptores de Activinas Tipo II/imunologia , Receptores de Activinas Tipo II/metabolismo , Adulto , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citocinas/imunologia , Citocinas/metabolismo , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Folistatina/imunologia , Folistatina/metabolismo , Folistatina/farmacologia , Humanos , Interleucina-13/farmacologia , Pessoa de Meia-Idade , Receptores de Fatores de Crescimento Transformadores beta/imunologia , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Proteína Smad2/imunologia , Proteína Smad2/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
18.
Cancer Res ; 80(16): 3359-3371, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32554750

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is a deadly and aggressive cancer. Understanding mechanisms that drive preneoplastic pancreatic lesions is necessary to improve early diagnostic and therapeutic strategies. Mutations and inactivation of activin-like kinase (ALK4) have been demonstrated to favor PDAC onset. Surprisingly, little is known regarding the ligands that drive ALK4 signaling in pancreatic cancer or how this signaling pathway limits the initiation of neoplastic lesions. In this study, data mining and histologic analyses performed on human and mouse tumor tissues revealed that activin A is the major ALK4 ligand that drives PDAC initiation. Activin A, which is absent in normal acinar cells, was strongly induced during acinar-to-ductal metaplasia (ADM), which was promoted by pancreatitis or the activation of KrasG12D in mice. Activin A expression during ADM was associated with the cellular senescence program that is induced in precursor lesions. Blocking activin A signaling through the use of a soluble form of activin receptor IIB (sActRIIB-Fc) and ALK4 knockout in mice expressing KrasG12D resulted in reduced senescence associated with decreased expression of p21, reduced phosphorylation of H2A histone family member X (H2AX), and increased proliferation. Thus, this study indicates that activin A acts as a protective senescence-associated secretory phenotype factor produced by Kras-induced senescent cells during ADM, which limits the expansion and proliferation of pancreatic neoplastic lesions. SIGNIFICANCE: This study identifies activin A to be a beneficial, senescence-secreted factor induced in pancreatic preneoplastic lesions, which limits their proliferation and ultimately slows progression into pancreatic cancers.


Assuntos
Receptores de Ativinas Tipo I/metabolismo , Ativinas/biossíntese , Carcinoma Ductal Pancreático/etiologia , Senescência Celular/fisiologia , Neoplasias Pancreáticas/etiologia , Lesões Pré-Cancerosas/etiologia , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II/metabolismo , Ativinas/antagonistas & inibidores , Animais , Carcinoma Ductal Pancreático/metabolismo , Progressão da Doença , Genes ras , Humanos , Camundongos , Neoplasias Pancreáticas/metabolismo , Fosforilação , Lesões Pré-Cancerosas/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Ativação Transcricional
19.
Int J Dev Biol ; 52(4): 353-63, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18415935

RESUMO

Various types of feeder cells have been adopted for the culture of human embryonic stem cells (hESCs) to improve their attachment and provide them with stemness-supporting factors. However, feeder cells differ in their capacity to support the growth of undifferentiated hESCs. Here, we compared the expression and secretion of four well-established regulators of hESC pluripotency and/or differentiation among five lines of human foreskin fibroblasts and primary mouse embryonic fibroblasts throughout a standard hESC culture procedure. We found that human and mouse feeder cells secreted comparable levels of TGF beta 1. However, mouse feeder cells secreted larger quantities of activin A than human feeder cells. Conversely, FGF-2, which was produced by human feeder cells, could not be detected in culture media from mouse feeder cells. The quantity of BMP-4 was at about the level of detectability in media from all feeder cell types, although BMP-4 dimers were present in all feeder cells. Production of TGF beta 1, activin A, and FGF-2 varied considerably among the human-derived feeder cell lines. Low- and high-producing human feeder cells as well as mouse feeder cells were evaluated for their ability to support the undifferentiated growth of hESCs. We found that a significantly lower proportion of hESCs maintained on human feeder cell types expressed SSEA3, an undifferentiated cell marker. Moreover, SSEA3 expression and thus the pluripotent hESC compartment could be partially rescued by addition of activin A. Cumulatively, these results suggest that the ability of a feeder layer to promote the undifferentiated growth of hESCs is attributable to its characteristic growth factor production.


Assuntos
Técnicas de Cocultura/métodos , Células-Tronco Embrionárias/citologia , Ativinas/biossíntese , Ativinas/genética , Animais , Antígenos Glicosídicos Associados a Tumores/genética , Sequência de Bases , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/análise , Proteínas Morfogenéticas Ósseas/genética , Diferenciação Celular , Proliferação de Células , Meios de Cultivo Condicionados/análise , Primers do DNA/genética , Células-Tronco Embrionárias/metabolismo , Fator 2 de Crescimento de Fibroblastos/biossíntese , Fator 2 de Crescimento de Fibroblastos/genética , Expressão Gênica , Glicoesfingolipídeos/genética , Humanos , Camundongos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Especificidade da Espécie , Antígenos Embrionários Estágio-Específicos , Fator de Crescimento Transformador beta1/biossíntese , Fator de Crescimento Transformador beta1/genética
20.
Arthritis Res Ther ; 21(1): 144, 2019 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-31182152

RESUMO

BACKGROUND: Activin A and follistatin exhibit immunomodulatory functions, thus affecting autoinflammatory processes as found in rheumatoid arthritis (RA). The impact of both proteins on the behavior of synovial fibroblasts (SF) in RA as well as in osteoarthritis (OA) is unknown. METHODS: Immunohistochemical analyses of synovial tissue for expression of activin A and follistatin were performed. The influence of RASF overexpressing activin A on cartilage invasion in a SCID mouse model was examined. RASF and OASF were stimulated with either IL-1ß or TNFα in combination with or solely with activin A, activin AB, or follistatin. Protein secretion was measured by ELISA and mRNA expression by RT-PCR. Smad signaling was confirmed by western blot. RESULTS: In human RA synovial tissue, the number of activin A-positive cells as well as its extracellular presence was higher than in the OA synovium. Single cells within the tissue expressed follistatin in RA and OA synovial tissue. In the SCID mouse model, activin A overexpression reduced RASF invasion. In human RASF, activin A was induced by IL-1ß and TNFα. Activin A slightly increased IL-6 release by unstimulated RASF, but decreased protein and mRNA levels of follistatin. CONCLUSION: The observed decrease of cartilage invasion by RASF overexpressing activin A in the SCID mouse model appears to be mediated by an interaction between activin/follistatin and other local cells indirectly affecting RASF because activin A displayed certain pro-inflammatory effects on RASF. Activin A even inhibits production and release of follistatin in RASF and therefore prevents itself from being blocked by its inhibitory binding protein follistatin in the local inflammatory joint environment.


Assuntos
Ativinas/genética , Artrite Reumatoide/genética , Folistatina/genética , Regulação da Expressão Gênica , Membrana Sinovial/metabolismo , Ativinas/biossíntese , Animais , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Fibroblastos/metabolismo , Fibroblastos/patologia , Folistatina/biossíntese , Humanos , Imuno-Histoquímica , Camundongos , Camundongos SCID , RNA/genética , Membrana Sinovial/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA