Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Int Heart J ; 62(5): 1135-1144, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34588407

RESUMO

Myocardial fibrosis is an important pathological phenomenon of cardiac remodeling that is induced by hypertension, myocardial ischemia, valvular heart disease, hypertrophic cardiomyopathy, and other heart diseases and can progress to heart failure. Urotensin II (UII) is regarded as a cardiovascular autacoid/hormone that is not only the most potent vasoconstrictor in mammals but also involved in cardiac remodeling. However, the molecular mechanisms responsible for UII-induced cardiac fibrosis have not yet been fully elucidated. Therefore, we aimed to investigate the effect of UII on myocardial fibrosis in cardiac hypertrophy and the mechanism of UII-induced cardiac fibrosis. Cardiac tissue from mice subjected to Transverse aortic constriction (TAC) was collected. Cardiac hypertrophy, myocardial fibrosis, and the expression of UII protein were assessed using echocardiography and pathological and molecular biological analyses. The effect of UII on fibrosis was evaluated in UII-treated mice and isolated rat primary cardiac fibroblasts, and the results indicated that UII induced significant myocardial fibrosis and increases in the proliferation and fibrotic responses both in mice and cultured fibroblasts. Mechanistically, UII treatment induced activation of the TGF-ß/Smad signaling pathway, which was suppressed by the UII receptor antagonist. In conclusion, UII plays critical roles in cardiac fibrosis by modulating the TGF-ß/Smads signaling pathway, which may be a promising therapeutic target in hypertrophic cardiomyopathy and related problems, such as cardiac remodeling and heart failure.


Assuntos
Cardiomegalia/etiologia , Miocárdio/patologia , Transdução de Sinais , Proteína Smad1/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Urotensinas/efeitos adversos , Animais , Fibrose/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
FASEB J ; 35(3): e21386, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33565137

RESUMO

Bone Morphogenetic Protein (BMP) signaling regulates diverse biological processes. Upon ligand binding, BMP receptors (BMPRs) phosphorylate SMAD1/5 and other noncanonical downstream effectors to induce transcription of downstream targets. However, the precise role of individual BMP receptors in this process remains largely unknown due to the complexity of downstream signaling and the innate promiscuity of ligand-receptor interaction. To delineate unique downstream effectors of individual BMPR1s, we analyzed the transcriptome of human umbilical endothelial cells (HUVECs) expressing three distinct constitutively active BMPR1s of which expression was detected in endothelial cells (ECs). From our analyses, we identified a number of novel downstream targets of BMPR1s in ECs. More importantly, we found that each BMPR1 possesses a distinctive set of downstream effectors, suggesting that each BMPR1 is likely to retain unique function in ECs. Taken together, our analyses suggest that each BMPR1 regulates downstream targets non-redundantly in ECs to create context-dependent outcomes of the BMP signaling.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/fisiologia , Perfilação da Expressão Gênica/métodos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Receptores de Ativinas Tipo I/genética , Animais , Células Cultivadas , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Smad1/fisiologia , Proteína Smad5/fisiologia
3.
Braz Oral Res ; 34: e006, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32022225

RESUMO

Induced pluripotent stem (iPS) cells could be induced into ameloblast-like cells by ameloblasts serum-free conditioned medium (ASF-CM), and bone morphogenetic proteins (BMPs) might be essential during the regulation of this process. The present study investigates the signal transduction that regulates the ameloblastic differentiation of iPS cells induced by ASF-CM. Mouse iPS cells were characterized and then cultured for 14 days in epithelial cell medium (control) or ASF-CM. Bone morphogenetic protein receptor II (BMPR-II) siRNA, inhibitor of Smad1/5 phosphorylation activated by activin receptor-like kinase (ALK) receptors, and inhibitors of mitogen-activated protein kinases (MAPKs) phosphorylation were used to treat the iPS cells in combination with ASF-CM. Real-time PCR, western blotting, and immunofluorescent staining were used to evaluate the expressions of ameloblast markers ameloblastin, enamelin, and cytokeratin-14. BMPR-II gene and protein levels increased markedly in ASF-CM-treated iPS cells compared with the controls, while the mRNA levels of Bmpr-Ia and Bmpr-Ib were similar between the ASF-CM and control groups. ASF-CM stimulation significantly increased the gene and protein expression of ameloblastin, enamelin and cytokeratin-14, and phosphorylated SMAD1/5, p38 MAPK, and ERK1/2 MAPK compared with the controls. Knockdown of BMPR-II and inhibition of Smad1/5 phosphorylation both could significantly reverse the increased expression of ameloblastin, enamelin, and cytokeratin-14 induced by ASF-CM, while neither inhibition of p38 nor ERK1/2 phosphorylation had significant reversing effects. We conclude that smad1/5 signaling transduction, activated by ALK receptors, regulates the ameloblastic differentiation of iPS cells induced by ameloblast-conditioned medium.


Assuntos
Ameloblastos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Transdução de Sinais/fisiologia , Proteína Smad1/fisiologia , Receptores de Ativinas/análise , Receptores de Ativinas/fisiologia , Western Blotting , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/análise , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Meios de Cultura Livres de Soro , Imunofluorescência , Expressão Gênica , Sistema de Sinalização das MAP Quinases/fisiologia , Fosforilação , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Smad1/análise , Fatores de Tempo , Proteínas Quinases p38 Ativadas por Mitógeno/análise , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
4.
Braz. oral res. (Online) ; 34: e006, 2020. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1089380

RESUMO

Abstract Induced pluripotent stem (iPS) cells could be induced into ameloblast-like cells by ameloblasts serum-free conditioned medium (ASF-CM), and bone morphogenetic proteins (BMPs) might be essential during the regulation of this process. The present study investigates the signal transduction that regulates the ameloblastic differentiation of iPS cells induced by ASF-CM. Mouse iPS cells were characterized and then cultured for 14 days in epithelial cell medium (control) or ASF-CM. Bone morphogenetic protein receptor II (BMPR-II) siRNA, inhibitor of Smad1/5 phosphorylation activated by activin receptor-like kinase (ALK) receptors, and inhibitors of mitogen-activated protein kinases (MAPKs) phosphorylation were used to treat the iPS cells in combination with ASF-CM. Real-time PCR, western blotting, and immunofluorescent staining were used to evaluate the expressions of ameloblast markers ameloblastin, enamelin, and cytokeratin-14. BMPR-II gene and protein levels increased markedly in ASF-CM-treated iPS cells compared with the controls, while the mRNA levels of Bmpr-Ia and Bmpr-Ib were similar between the ASF-CM and control groups. ASF-CM stimulation significantly increased the gene and protein expression of ameloblastin, enamelin and cytokeratin-14, and phosphorylated SMAD1/5, p38 MAPK, and ERK1/2 MAPK compared with the controls. Knockdown of BMPR-II and inhibition of Smad1/5 phosphorylation both could significantly reverse the increased expression of ameloblastin, enamelin, and cytokeratin-14 induced by ASF-CM, while neither inhibition of p38 nor ERK1/2 phosphorylation had significant reversing effects. We conclude that smad1/5 signaling transduction, activated by ALK receptors, regulates the ameloblastic differentiation of iPS cells induced by ameloblast-conditioned medium.


Assuntos
Transdução de Sinais/fisiologia , Proteína Smad1/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Ameloblastos/citologia , Fosforilação , Fatores de Tempo , Expressão Gênica , Diferenciação Celular/fisiologia , Diferenciação Celular/genética , Células Cultivadas , Western Blotting , Imunofluorescência , Meios de Cultura Livres de Soro , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sistema de Sinalização das MAP Quinases/fisiologia , Receptores de Ativinas/análise , Receptores de Ativinas/fisiologia , Interferência de RNA , Proteínas Quinases p38 Ativadas por Mitógeno/análise , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/análise , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Proteína Smad1/análise
5.
Braz. oral res. (Online) ; 34: e006, 2020. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1055522

RESUMO

Abstract Induced pluripotent stem (iPS) cells could be induced into ameloblast-like cells by ameloblasts serum-free conditioned medium (ASF-CM), and bone morphogenetic proteins (BMPs) might be essential during the regulation of this process. The present study investigates the signal transduction that regulates the ameloblastic differentiation of iPS cells induced by ASF-CM. Mouse iPS cells were characterized and then cultured for 14 days in epithelial cell medium (control) or ASF-CM. Bone morphogenetic protein receptor II (BMPR-II) siRNA, inhibitor of Smad1/5 phosphorylation activated by activin receptor-like kinase (ALK) receptors, and inhibitors of mitogen-activated protein kinases (MAPKs) phosphorylation were used to treat the iPS cells in combination with ASF-CM. Real-time PCR, western blotting, and immunofluorescent staining were used to evaluate the expressions of ameloblast markers ameloblastin, enamelin, and cytokeratin-14. BMPR-II gene and protein levels increased markedly in ASF-CM-treated iPS cells compared with the controls, while the mRNA levels of Bmpr-Ia and Bmpr-Ib were similar between the ASF-CM and control groups. ASF-CM stimulation significantly increased the gene and protein expression of ameloblastin, enamelin and cytokeratin-14, and phosphorylated SMAD1/5, p38 MAPK, and ERK1/2 MAPK compared with the controls. Knockdown of BMPR-II and inhibition of Smad1/5 phosphorylation both could significantly reverse the increased expression of ameloblastin, enamelin, and cytokeratin-14 induced by ASF-CM, while neither inhibition of p38 nor ERK1/2 phosphorylation had significant reversing effects. We conclude that smad1/5 signaling transduction, activated by ALK receptors, regulates the ameloblastic differentiation of iPS cells induced by ameloblast-conditioned medium.


Assuntos
Transdução de Sinais/fisiologia , Proteína Smad1/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Ameloblastos/citologia , Fosforilação , Fatores de Tempo , Expressão Gênica , Diferenciação Celular/fisiologia , Diferenciação Celular/genética , Células Cultivadas , Western Blotting , Imunofluorescência , Meios de Cultura Livres de Soro , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sistema de Sinalização das MAP Quinases/fisiologia , Receptores de Ativinas/análise , Receptores de Ativinas/fisiologia , Interferência de RNA , Proteínas Quinases p38 Ativadas por Mitógeno/análise , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/análise , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Proteína Smad1/análise
6.
Hepatology ; 70(6): 1986-2002, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31127639

RESUMO

A failure of iron to appropriately regulate liver hepcidin production is central to the pathogenesis of hereditary hemochromatosis. SMAD1/5 transcription factors, activated by bone morphogenetic protein (BMP) signaling, are major regulators of hepcidin production in response to iron; however, the role of SMAD8 and the contribution of SMADs to hepcidin production by other systemic cues remain uncertain. Here, we generated hepatocyte Smad8 single (Smad8fl/fl ;Alb-Cre+ ), Smad1/5/8 triple (Smad158;Alb-Cre+ ), and littermate Smad1/5 double (Smad15;Alb-Cre+ ) knockout mice to investigate the role of SMAD8 in hepcidin and iron homeostasis regulation and liver injury. We found that Smad8;Alb-Cre+ mice exhibited no iron phenotype, whereas Smad158;Alb-Cre+ mice had greater iron overload than Smad15;Alb-Cre+ mice. In contrast to the sexual dimorphism reported for wild-type mice and other hemochromatosis models, hepcidin deficiency and extrahepatic iron loading were similarly severe in Smad15;Alb-Cre+ and Smad158;Alb-Cre+ female compared with male mice. Moreover, epidermal growth factor (EGF) failed to suppress hepcidin in Smad15;Alb-Cre+ hepatocytes. Conversely, hepcidin was still increased by lipopolysaccharide in Smad158;Alb-Cre+ mice, although lower basal hepcidin resulted in lower maximal hepcidin. Finally, unlike most mouse hemochromatosis models, Smad158;Alb-Cre+ developed liver injury and fibrosis at 8 weeks. Liver injury and fibrosis were prevented in Smad158;Alb-Cre+ mice by a low-iron diet and were minimal in iron-loaded Cre- mice. Conclusion: Hepatocyte Smad1/5/8 knockout mice are a model of hemochromatosis that encompasses liver injury and fibrosis seen in human disease. These mice reveal the redundant but critical role of SMAD8 in hepcidin and iron homeostasis regulation, establish a requirement for SMAD1/5/8 in hepcidin regulation by testosterone and EGF but not inflammation, and suggest a pathogenic role for both iron loading and SMAD1/5/8 deficiency in liver injury and fibrosis.


Assuntos
Hepatócitos/metabolismo , Sobrecarga de Ferro/etiologia , Ferro/metabolismo , Cirrose Hepática Experimental/etiologia , Proteína Smad1/fisiologia , Proteína Smad5/fisiologia , Proteína Smad8/fisiologia , Animais , Células Cultivadas , Fator de Crescimento Epidérmico/farmacologia , Feminino , Hepcidinas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
7.
Life Sci ; 220: 106-116, 2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30708099

RESUMO

AIMS: This study explored the role of the BMP4/Smad1 signaling pathway in mesangial matrix expansion during the process of diabetic nephropathy. MAIN METHODS: Diabetic rats were induced by high-fat feeding followed by an intraperitoneal injection of streptozotocin. Glomerular lesions were examined. Immunohistochemical analysis was performed in order to identify BMP4/Smad1 signaling proteins (BMP4, ALK3, and Smad1) and mesangial ECM proteins (Col1 and Col4) in kidney tissue. Cell proliferation and the expression of BMP4, Smad1 and Col4 were determined in cultured mesangial cells exposed to high glucose. The specific regulatory role of BMP4 was evaluated by detecting BMP4/Smad1 signaling pathway proteins and mesangial ECM proteins after blocking BMP4 both at the gene and protein levels. KEY FINDINGS: Rats with DN exhibited mesangial expansion and a thickened glomerular basement membrane. Immunohistochemical analysis of glomeruli showed increased expression of BMP4, Smad1, ALK3, Col1, and Col4 but less expression of MMP9 than observed in controls. High glucose induced slight proliferation of cultured rat mesangial cells after 48 h of incubation but there was no significant different from the control (p > 0.05). High glucose activated the BMP4/Smad1 signaling pathway and stimulated Col4 expression in mesangial cells. Both silencing of the bmp4 gene (with siRNA) and blocking BMP4 protein signaling (with the BMP4 protein antagonist Noggin) reduced the expression of ALK3, Smad1, Col4, and Col1 in high glucose-stimulated mesangial cells. SIGNIFICANCE: The BMP4/Smad1 signaling pathway is crucial to the progression of mesangial expansion, and suppressing this signaling pathway may present a novel therapeutic strategy for DN.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Nefropatias Diabéticas/metabolismo , Proteína Smad1/metabolismo , Animais , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/fisiologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/fisiologia , Proliferação de Células/fisiologia , Colágeno Tipo IV/metabolismo , Diabetes Mellitus Experimental/metabolismo , Modelos Animais de Doenças , Mesângio Glomerular/metabolismo , Rim/metabolismo , Glomérulos Renais/metabolismo , Masculino , Células Mesangiais/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Proteína Smad1/fisiologia , Estreptozocina/farmacologia
8.
Small GTPases ; 10(1): 26-32, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-28125332

RESUMO

In this commentary we discuss a paper we published recently on the activities of the GTPase RhoA during neural differentiation of murine embryonic stem cells, and relate our findings to previous studies. We narrate how we found that RhoA impedes neural differentiation by inhibiting the production as well as the secretion of noggin, a soluble factor that antagonizes bone morphogenetic protein. We discuss how the questions we tried to address shaped the study, and how embryonic stem cells isolated from a genetically modified mouse model devoid of Syx, a RhoA-specific guanine exchange factor, were used to address them. We detail several signaling pathways downstream of RhoA that are hindered by the absence of Syx, and obstructed by retinoic acid, resulting in an increase of noggin production; we explain how the lower RhoA activity and, consequently, the sparser peri-junctional stress fibers in Syx-/- cells facilitated noggin secretion; and we report unpublished results showing that pharmacological inhibition of RhoA accelerates the neuronal differentiation of human embryonic stem cells. Finally, we identify signaling mechanisms in our recent study that warrant further study, and speculate on the possibility of manipulating RhoA signaling in combination with other pathways to drive the differentiation of neuronal subtypes.


Assuntos
Células-Tronco Embrionárias/citologia , Neurogênese , Proteína rhoA de Ligação ao GTP/fisiologia , Animais , Proteínas de Transporte/fisiologia , Células-Tronco Embrionárias/fisiologia , Humanos , Proteína Smad1/fisiologia , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores
9.
Biol Psychiatry ; 84(12): 881-892, 2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30158054

RESUMO

BACKGROUND: Substance use disorder is a neurobiological disease characterized by episodes of relapse despite periods of withdrawal. It is thought that neuroadaptations in discrete brain areas of the reward pathway, including the nucleus accumbens, underlie these aberrant behaviors. The ubiquitin-proteasome system degrades proteins and has been shown to be involved in cocaine-induced plasticity, but the role of E3 ubiquitin ligases, which conjugate ubiquitin to substrates, is unknown. Here, we examined E3 ubiquitin-protein ligase SMURF1 (SMURF1) in neuroadaptations and relapse behavior during withdrawal following cocaine self-administration. METHODS: SMURF1 and downstream targets ras homolog gene family, member A (RhoA), SMAD1/5, and Runt-related transcript factor 2 were examined using Western blotting (n = 9-11/group), quantitative polymerase chain reaction (n = 6-9/group), co-immunoprecipitation (n = 9-11/group), tandem ubiquitin binding entities affinity purification (n = 5-6/group), and quantitative chromatin immunoprecipitation (n = 3-6/group) (2 rats/sample). Viral-mediated gene transfer (n = 7-12/group) and intra-accumbal microinjections (n = 9-10/group) were used to examine causal roles of SMURF1 and substrate RhoA, respectively, in cue-induced cocaine seeking. RESULTS: SMURF1 protein expression was decreased, while SMURF1 substrates RhoA and SMAD1/5 were increased, in the nucleus accumbens on withdrawal day 7, but not on withdrawal day 1, following cocaine self-administration. Viral-mediated gene transfer of Smurf1 or constitutive activation of RhoA attenuated cue-induced cocaine seeking, while catalytically inactive Smurf1 enhanced cocaine seeking. Furthermore, SMURF1-regulated, SMAD1/5-associated transcription factor Runt-related transcript factor 2 displayed increased binding at promoter regions of genes previously associated with cocaine-induced plasticity. CONCLUSIONS: SMURF1 is a key mediator of neuroadaptations in the nucleus accumbens following cocaine exposure and mediates cue-induced cocaine seeking during withdrawal.


Assuntos
Cocaína/administração & dosagem , Comportamento de Procura de Droga/fisiologia , Núcleo Accumbens/fisiologia , Proteína Smad1/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Animais , Transtornos Relacionados ao Uso de Cocaína/genética , Sinais (Psicologia) , Masculino , Núcleo Accumbens/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma , Ratos , Ratos Sprague-Dawley , Autoadministração , Transdução de Sinais , Proteína Smad1/genética , Ubiquitina-Proteína Ligases/genética
10.
Development ; 144(23): 4298-4312, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28870989

RESUMO

How position-dependent cell fate acquisition occurs during embryogenesis is a central question in developmental biology. To study this process, we developed a defined, high-throughput assay to induce peri-gastrulation-associated patterning in geometrically confined human pluripotent stem cell (hPSC) colonies. We observed that, upon BMP4 treatment, phosphorylated SMAD1 (pSMAD1) activity in the colonies organized into a radial gradient. We developed a reaction-diffusion (RD)-based computational model and observed that the self-organization of pSMAD1 signaling was consistent with the RD principle. Consequent fate acquisition occurred as a function of both pSMAD1 signaling strength and duration of induction, consistent with the positional-information (PI) paradigm. We propose that the self-organized peri-gastrulation-like fate patterning in BMP4-treated geometrically confined hPSC colonies arises via a stepwise model of RD followed by PI. This two-step model predicted experimental responses to perturbations of key parameters such as colony size and BMP4 dose. Furthermore, it also predicted experimental conditions that resulted in RD-like periodic patterning in large hPSC colonies, and rescued peri-gastrulation-like patterning in colony sizes previously thought to be reticent to this behavior.


Assuntos
Padronização Corporal/fisiologia , Gastrulação/fisiologia , Modelos Biológicos , Padronização Corporal/genética , Proteína Morfogenética Óssea 4/fisiologia , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias/métodos , Gastrulação/genética , Ensaios de Triagem em Larga Escala/métodos , Humanos , Proteína Nodal/fisiologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , RNA Interferente Pequeno/genética , Transdução de Sinais , Proteína Smad1/fisiologia
11.
Naunyn Schmiedebergs Arch Pharmacol ; 390(7): 711-720, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28374099

RESUMO

Reactive oxygen species (ROS) is a pivotal pathogenic factor in the development of osteoporosis. Dalbergioidin (DAL) can be isolated from Uraria crinite, an edible herb used as a natural food for childhood skeletal dysplasia. Recent research has implicated DAL as having an antiosteoporosis effect, although the mechanism of this is unclear. We used an effective oxidative stress model, induced by hydrogen peroxide (H2O2) in osteoblastic MC3T3-E1 cells, to investigate the protective effects of DAL in osteoporosis and the underlying molecular mechanisms. The results indicated that treatment with DAL maintained redox balance, reduced MC3T3-E1 cell apoptosis, improved alkaline phosphatase activity, and elevated the osteogenic-related protein expression of Runx2, Osterix, and BMP2 against oxidative damage induced by H2O2. The potential molecular mechanism involved in the protective effect of DAL against H2O2-induced cell death in MC3T3-E1 cells may lie in the activation of the PI3K/AKT/SMAD1 cell signal pathway. Taken together, the results indicated that the potential protective effects of DAL against osteoporosis were linked to a reduction in oxidative damage, suggesting that DAL may be useful in bone metabolism diseases, particularly osteoporosis.


Assuntos
Cromonas/farmacologia , Peróxido de Hidrogênio/toxicidade , Osteoblastos/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/fisiologia , Fosfatase Alcalina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Camundongos , Osteoporose/tratamento farmacológico
12.
Reproduction ; 153(2): 211-220, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27864336

RESUMO

Generation of male germ cells from pluripotent cells could provide male gametes for treating male infertility and offer an ideal model for unveiling molecular mechanisms of spermatogenesis. However, the influence and exact molecular mechanisms, especially downstream effectors of BMP4 signaling pathways, in male germ cell differentiation of the induce pluripotent stem (iPS) cells, remain unknown. This study was designed to explore the role and mechanism of BMP4 signaling in the differentiation of mouse iPS cells to male germ cells. Embryoid body (EB) formation and recombinant BMP4 or Noggin were utilized to evaluate the effect of BMP4 on male germ cell generation from mouse iPS cells. Germ cell-specific genes and proteins as well as the downstream effectors of BMP4 signaling pathway were assessed using real-time PCR and Western blots. We found that BMP4 ligand and its multiple receptors, including BMPR1a, BMPR1b and BMPR2, were expressed in mouse iPS cells. Real-time PCR and Western blots revealed that BMP4 could upregulate the levels of genes and proteins for germ cell markers in iPS cells-derived EBs, whereas Noggin decreased their expression in these cells. Moreover, Smad1/5 phosphorylation, Gata4 transcription and the transcripts of Id1 and Id2 were enhanced by BMP4 but decreased when exposed to Noggin. Collectively, these results suggest that BMP4 promotes the generation of male germ cells from iPS cells via Smad1/5 pathway and the activation of Gata4, Id1 and Id2 This study thus offers novel insights into molecular mechanisms underlying male germ cell development.


Assuntos
Proteína Morfogenética Óssea 4/fisiologia , Diferenciação Celular/fisiologia , Células Germinativas/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Western Blotting , Proteína Morfogenética Óssea 4/genética , Linhagem Celular , Fator de Transcrição GATA4/fisiologia , Expressão Gênica , Células-Tronco Pluripotentes Induzidas/fisiologia , Proteína 1 Inibidora de Diferenciação/fisiologia , Proteína 2 Inibidora de Diferenciação/fisiologia , Masculino , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia , Proteína Smad1/fisiologia , Proteína Smad5/fisiologia , Espermatozoides/citologia
13.
Dev Biol ; 410(1): 98-107, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26681395

RESUMO

Nedd4 is an E3 ubiquitin ligase that has an essential role in craniofacial development. However, how and when Nedd4 controls skull formation is ill defined. Here we have used a collection of complementary genetic mouse models to dissect the cell-autonomous roles of Nedd4 in the formation of neural crest cell derived cranial bone. Removal of Nedd4 specifically from neural crest cells leads to profound craniofacial defects with marked reduction of cranial bone that was preceded by hypoplasia of bone forming osteoblasts. Removal of Nedd4 after differentiation of neural crest cells into progenitors of chondrocytes and osteoblasts also led to profound deficiency of craniofacial bone in the absence of cartilage defects. Notably, these skull malformations were conserved when Nedd4 was specifically removed from the osteoblast lineage after specification of osteoblast precursors from mesenchymal skeletal progenitors. We further show that absence of Nedd4 in pre-osteoblasts results in decreased cell proliferation and altered osteogenic differentiation. Taken together our data demonstrate a novel cell-autonomous role for Nedd4 in promoting expansion of the osteoblast progenitor pool to control craniofacial development. Nedd4 mutant mice therefore represent a unique mouse model of craniofacial anomalies that provide an ideal resource to explore the cell-intrinsic mechanisms of neural crest cells in craniofacial morphogenesis.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/fisiologia , Ossos Faciais/embriologia , Osteogênese , Crânio/embriologia , Ubiquitina-Proteína Ligases/fisiologia , Animais , Linhagem da Célula , Proliferação de Células , Camundongos , Ubiquitina-Proteína Ligases Nedd4 , Crista Neural/fisiologia , Osteoblastos/citologia , Proteína Smad1/fisiologia
14.
Arch Oral Biol ; 60(9): 1319-26, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26123746

RESUMO

OBJECTIVE: BMP-2 induces osteoblast differentiation and activates osteoclast formation. Here, we investigated the role of Smad1, a molecule that signals downstream of BMP-2, in mediating the effects of BMP-2 on osteoclast differentiation induced by 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) in osteoblasts. DESIGN: The effects of 1,25(OH)2D3 and BMP-2 in osteoclasts were examined using polymerase chain reaction and Western blotting to measure changes in target gene and protein expression. Immunostaining was carried out to investigate the localization of the vitamin D receptor (VDR) in the nucleus in response to BMP-2. RESULTS: Stimulation with both 1,25(OH)2D3 and BMP-2 resulted in significantly greater osteoclast formation and receptor activator of nuclear factor κB ligand (RANKL) mRNA expression compared to stimulation with 1,25(OH)2D3 alone. In addition, expression of the VDR protein was increased, enhancing the activity of 1,25(OH)2D3. Interestingly, knockdown of Smad1 resulted in reduced osteoclast formation, RANKL mRNA expression, and VDR protein expression compared with control cells. Costimulation with 1,25(OH)2D3 and BMP-2 enhanced VDR localization in the nucleus. CONCLUSIONS: We found that BMP-2 induced Smad1 activation, thereby influencing the localization of VDR in the nucleus in the presence of 1,25(OH)2D3 and resulting in increased RANKL mRNA expression. These effects ultimately resulted in enhanced osteoclast differentiation.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Interferência de RNA , Proteína Smad1/fisiologia , Animais , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Ligantes , Camundongos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Reação em Cadeia da Polimerase , RNA Mensageiro/metabolismo , Receptor Ativador de Fator Nuclear kappa-B , Receptores de Calcitriol/metabolismo , Transdução de Sinais
15.
Cardiovasc Res ; 107(1): 108-18, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25824146

RESUMO

AIMS: Previously we demonstrated that both hypoxia inducible factor-1 (HIF-1) and bone morphogenetic protein-4 (BMP4) up-regulate transient receptor potential canonical (TRPC) 1 and TRPC6, resulting in increased basal intracellular Ca(2+) concentration ([Ca(2+)]i) in pulmonary arterial smooth muscle cells (PASMCs), driving development of chronic hypoxia (CH)-induced pulmonary hypertension (CHPH). This study aims to determine whether HIF-1 regulates BMP4, and whether BMP4 mediates TRPC and basal [Ca(2+)]i increases in hypoxic PASMCs. METHODS AND RESULTS: The level of BMP4 mature protein was increased for ∼183% in distal pulmonary arterial smooth muscle (PA) from CH (10% O2 for 21 days; CH) exposed rats, and 143% in PASMCs cultured under prolonged hypoxia (4% O2 for 60 h). In rat PASMCs, HIF-1α overexpression up-regulated, whereas HIF-1α knockdown under hypoxia decreased BMP4 expression; site-mutation identified two functional HIF-1-binding sites in Bmp4 gene promoter; noggin or BMP4 siRNA treatment blocked hypoxia-induced increases of TRPC1 and TRPC6 expression and basal [Ca(2+)]i. Likewise, in mice, exposure to CH increased BMP4 expression in distal PA for ∼80%, which was absent in HIF-1α heterozygous mutant mice. Comparing with wild-type littermates, BMP4 heterozygous mutant mice exposed to CH displayed lower BMP4 and TRPC levels in PA, decreased basal [Ca(2+)]i in PASMCs, and attenuated CHPH. In human PASMCs, HIF-1α knockdown attenuated hypoxia-induced BMP4 expression and knockdown of either HIF-1α or BMP4 abolished hypoxia-induced TRPC expression and basal [Ca(2+)]i. CONCLUSIONS: BMP4 acts downstream of HIF-1 and mediates hypoxia-induced up-regulation of TRPC, leading to increased basal [Ca(2+)]i in PASMCs, promoting CHPH pathogenesis.


Assuntos
Proteína Morfogenética Óssea 4/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Hipóxia/metabolismo , Artéria Pulmonar/metabolismo , Canais de Cátion TRPC/genética , Animais , Proteína Morfogenética Óssea 4/genética , Cálcio/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Hipertensão Pulmonar/etiologia , Camundongos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Ratos , Proteína Smad1/fisiologia , Canal de Cátion TRPC6 , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Dev Biol ; 397(1): 45-55, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25446538

RESUMO

The mechanisms regulating cell division during development of the mouse pre-implantation embryo are poorly understood. We have investigated whether bone morphogenetic protein (BMP) signaling is involved in controlling cell cycle during mouse pre-implantation development. We mapped and quantitated the dynamic activities of BMP signaling through high-resolution immunofluorescence imaging combined with a 3D segmentation method. Immunostaining for phosphorylated Smad1/5/8 shows that BMP signaling is activated in mouse embryos as early as the 4-cell stage, and becomes spatially restricted by late blastocyst stage. Perturbation of BMP signaling in preimplantation mouse embryos, whether by treatment with a small molecule inhibitor, with Noggin protein, or by overexpression of a dominant-negative BMP receptor, indicates that BMPs regulate cell cleavage up to the morula stage. These results indicate that BMP signaling is active during mouse pre-implantation development and is required for cell cleavage in preimplantation mouse embryos.


Assuntos
Blastocisto/fisiologia , Proteínas Morfogenéticas Ósseas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Animais , Proteínas de Transporte/metabolismo , Divisão Celular , Células Cultivadas , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias/citologia , Feminino , Genes Dominantes , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Microscopia de Fluorescência , Fosforilação , Transdução de Sinais , Proteína Smad1/fisiologia , Proteína Smad5/fisiologia , Proteína Smad8/fisiologia , Fatores de Tempo
17.
Endocrinology ; 155(8): 3054-64, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24848867

RESUMO

TGFß superfamily proteins, acting via SMAD (Sma- and Mad-related protein)2/3 pathways, regulate placental function; however, the role of SMAD1/5/8 pathway in the placenta is unknown. This study investigated the functional role of bone morphogenetic protein (BMP)4 signaling through SMAD1/5 in terminal differentiation of primary chorionic gonadotropin (CG)-secreting trophoblast. Primary equine trophoblast cells or placental tissues were isolated from day 27-34 equine conceptuses. Detected by microarray, RT-PCR, and quantitative RT-PCR, equine chorionic girdle trophoblast showed increased gene expression of receptors that bind BMP4. BMP4 mRNA expression was 20- to 60-fold higher in placental tissues adjacent to the chorionic girdle compared with chorionic girdle itself, suggesting BMP4 acts primarily in a paracrine manner on the chorionic girdle. Stimulation of chorionic girdle-trophoblast cells with BMP4 resulted in a dose-dependent and developmental stage-dependent increase in total number and proportion of terminally differentiated binucleate cells. Furthermore, BMP4 treatment induced non-CG-secreting day 31 chorionic girdle trophoblast cells to secrete CG, confirming a specific functional response to BMP4 stimulation. Inhibition of SMAD2/3 signaling combined with BMP4 treatment further enhanced differentiation of trophoblast cells. Phospho-SMAD1/5, but not phospho-SMAD2, expression as determined by Western blotting was tightly regulated during chorionic girdle trophoblast differentiation in vivo, with peak expression of phospho-SMAD1/5 in vivo noted at day 31 corresponding to maximal differentiation response of trophoblast in vitro. Collectively, these experiments demonstrate the involvement of BMP4-dependent pathways in the regulation of equine trophoblast differentiation in vivo and primary trophoblast differentiation in vitro via activation of SMAD1/5 pathway, a previously unreported mechanism of TGFß signaling in the mammalian placenta.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Diferenciação Celular , Gonadotropina Coriônica/metabolismo , Proteínas Smad Reguladas por Receptor/metabolismo , Trofoblastos/citologia , Animais , Feminino , Cavalos , Gravidez , Cultura Primária de Células , Transdução de Sinais/fisiologia , Proteína Smad1/fisiologia , Proteína Smad5/fisiologia , Proteína Smad8/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Trofoblastos/metabolismo
18.
Proc Natl Acad Sci U S A ; 111(20): 7337-42, 2014 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-24808138

RESUMO

In vertebrate embryos, cardiac progenitor cells (CPCs) undergo long-range migration after emerging from the primitive streak during gastrulation. Together with other mesoderm progenitors, they migrate laterally and then toward the ventral midline, where they form the heart. Signals controlling the migration of different progenitor cell populations during gastrulation are poorly understood. Several pathways are involved in the epithelial-to-mesenchymal transition and ingression of mesoderm cells through the primitive streak, including fibroblast growth factors and wingless-type family members (Wnt). Here we focus on early CPC migration and use live video microscopy in chicken embryos to demonstrate a role for bone morphogenetic protein (BMP)/SMA and MAD related (Smad) signaling. We identify an interaction of BMP and Wnt/glycogen synthase kinase 3 beta (GSK3ß) pathways via the differential phosphorylation of Smad1. Increased BMP2 activity altered migration trajectories of prospective cardiac cells and resulted in their lateral displacement and ectopic differentiation, as they failed to reach the ventral midline. Constitutively active BMP receptors or constitutively active Smad1 mimicked this phenotype, suggesting a cell autonomous response. Expression of GSK3ß, which promotes the turnover of active Smad1, rescued the BMP-induced migration phenotype. Conversely, expression of GSK3ß-resistant Smad1 resulted in aberrant CPC migration trajectories. De-repression of GSK3ß by dominant negative Wnt3a restored normal migration patterns in the presence of high BMP activity. The data indicate the convergence of BMP and Wnt pathways on Smad1 during the early migration of prospective cardiac cells. Overall, we reveal molecular mechanisms that contribute to the emerging paradigm of signaling pathway integration in embryo development.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Miocárdio/citologia , Miocárdio/metabolismo , Proteína Smad1/fisiologia , Células-Tronco/citologia , Proteína Wnt3A/metabolismo , Animais , Padronização Corporal , Diferenciação Celular , Movimento Celular , Embrião de Galinha , Genes Dominantes , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Coração/embriologia , Mesoderma/metabolismo , Fenótipo , Linha Primitiva/metabolismo , Transdução de Sinais
19.
Prostate ; 74(2): 121-33, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24185914

RESUMO

INTRODUCTION. Overexpression of bone morphogenetic protein-6 (BMP-6) has been reported in human prostate cancer tissues. Previously we have demonstrated that BMP-6 enhances prostate cancer growth in mice and not in tissue culture. Herein, we have investigated the mechanism of BMP-6's pro-tumorigenic effect in prostate cancer. METHODS. Tramp C2 murine and LNCaP human prostate cancer cell lines were co-cultured with RAW 264.7 and THP-1 cells, respectively. IL-1a knockout mice were used to confirm the role of BMP-6/IL-1a loop in vivo. Lastly, conditional macrophage null mice cd11b-DTR was used. RESULTS. The results demonstrated that BMP-6 induced the expression of IL-1a in macrophages via a cross-talk between NF-kB1 p50 and Smad1. When endothelial cells were treated with conditioned media harvested from macrophages incubated with BMP-6, tube formation was detected. In the presence of IL-1a neutralizing antibody, endothelial tube formation was blocked. In vivo, tumor growth and neovascularization decreased significantly when BMP-6 was expressed in IL-1a knockout and conditional macrophage-null mice. CONCLUSIONS. Prostate cancer-derived BMP-6 stimulates tumor-associated macrophages to produce IL-1a through a crosstalk between Smad1 and NF-kB1; IL-1a, in turn, promotes angiogenesis and prostate cancer growth.


Assuntos
Proteína Morfogenética Óssea 6/fisiologia , Carcinogênese/patologia , Interleucina-1alfa/fisiologia , Macrófagos/patologia , Neovascularização Patológica/fisiopatologia , Neoplasias da Próstata/patologia , Animais , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Endotélio Vascular/patologia , Humanos , Interleucina-1alfa/deficiência , Interleucina-1alfa/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/fisiologia , Neoplasias da Próstata/irrigação sanguínea , Transdução de Sinais/fisiologia , Proteína Smad1/fisiologia
20.
Circ Res ; 113(11): 1231-41, 2013 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-24047927

RESUMO

RATIONALE: The rapid induction and orchestration of new blood vessels are critical for tissue repair in response to injury, such as myocardial infarction, and for physiological angiogenic responses, such as embryonic development and exercise. OBJECTIVE: We aimed to identify and characterize microRNAs (miR) that regulate pathological and physiological angiogenesis. METHODS AND RESULTS: We show that miR-26a regulates pathological and physiological angiogenesis by targeting endothelial cell (EC) bone morphogenic protein/SMAD1 signaling in vitro and in vivo. MiR-26a expression is increased in a model of acute myocardial infarction in mice and in human subjects with acute coronary syndromes. Ectopic expression of miR-26a markedly induced EC cycle arrest and inhibited EC migration, sprouting angiogenesis, and network tube formation in matrigel, whereas blockade of miR-26a had the opposite effects. Mechanistic studies demonstrate that miR-26a inhibits the bone morphogenic protein/SMAD1 signaling pathway in ECs by binding to the SMAD1 3'-untranslated region, an effect that decreased expression of Id1 and increased p21(WAF/CIP) and p27. In zebrafish, miR-26a overexpression inhibited formation of the caudal vein plexus, a bone morphogenic protein-responsive process, an effect rescued by ectopic SMAD1 expression. In mice, miR-26a overexpression inhibited EC SMAD1 expression and exercise-induced angiogenesis. Furthermore, systemic intravenous administration of an miR-26a inhibitor, locked nucleic acid-anti-miR-26a, increased SMAD1 expression and rapidly induced robust angiogenesis within 2 days, an effect associated with reduced myocardial infarct size and improved heart function. CONCLUSIONS: These findings establish miR-26a as a regulator of bone morphogenic protein/SMAD1-mediated EC angiogenic responses, and that manipulating miR-26a expression could provide a new target for rapid angiogenic therapy in ischemic disease states.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , MicroRNAs/fisiologia , Neovascularização Patológica/fisiopatologia , Neovascularização Fisiológica/fisiologia , Transdução de Sinais/fisiologia , Proteína Smad1/fisiologia , Síndrome Coronariana Aguda/sangue , Síndrome Coronariana Aguda/patologia , Síndrome Coronariana Aguda/fisiopatologia , Animais , Biomarcadores/sangue , Proliferação de Células , Modelos Animais de Doenças , Desenvolvimento Embrionário/fisiologia , Endotélio Vascular/patologia , Endotélio Vascular/fisiologia , Endotélio Vascular/fisiopatologia , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/sangue , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda/fisiologia , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...