Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Elife ; 122024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38856718

RESUMO

Abnormal lung development can cause congenital pulmonary cysts, the mechanisms of which remain largely unknown. Although the cystic lesions are believed to result directly from disrupted airway epithelial cell growth, the extent to which developmental defects in lung mesenchymal cells contribute to abnormal airway epithelial cell growth and subsequent cystic lesions has not been thoroughly examined. In the present study using genetic mouse models, we dissected the roles of bone morphogenetic protein (BMP) receptor 1a (Bmpr1a)-mediated BMP signaling in lung mesenchyme during prenatal lung development and discovered that abrogation of mesenchymal Bmpr1a disrupted normal lung branching morphogenesis, leading to the formation of prenatal pulmonary cystic lesions. Severe deficiency of airway smooth muscle cells and subepithelial elastin fibers were found in the cystic airways of the mesenchymal Bmpr1a knockout lungs. In addition, ectopic mesenchymal expression of BMP ligands and airway epithelial perturbation of the Sox2-Sox9 proximal-distal axis were detected in the mesenchymal Bmpr1a knockout lungs. However, deletion of Smad1/5, two major BMP signaling downstream effectors, from the lung mesenchyme did not phenocopy the cystic abnormalities observed in the mesenchymal Bmpr1a knockout lungs, suggesting that a Smad-independent mechanism contributes to prenatal pulmonary cystic lesions. These findings reveal for the first time the role of mesenchymal BMP signaling in lung development and a potential pathogenic mechanism underlying congenital pulmonary cysts.


Congenital disorders are medical conditions that are present from birth. Although many congenital disorders are rare, they can have a severe impact on the quality of life of those affected. For example, congenital pulmonary airway malformation (CPAM) is a rare congenital disorder that occurs in around 1 out of every 25,000 pregnancies. In CPAM, abnormal, fluid-filled sac-like pockets of tissue, known as cysts, form within the lungs of unborn babies. After birth, these cysts become air-filled and do not behave like normal lung tissue and stop a baby's lungs from working properly. In severe cases, babies with CPAM need surgery immediately after birth. We still do not understand exactly what the underlying causes of CPAM might be. CPAM is not considered to be hereditary ­ that is, it does not appear to be passed down in families ­ nor is it obviously linked to any environmental factors. CPAM is also very difficult to study, because researchers cannot access tissue samples during the critical early stages of the disease. To overcome these difficulties, Luo et al. wanted to find a way to study CPAM in the laboratory. First, they developed a non-human animal 'model' that naturally forms CPAM-like lung cysts, using genetically modified mice where the gene for the signaling molecule Bmpr1a had been deleted in lung cells. Normally, Bmpr1a is part of a set of the molecular instructions, collectively termed BMP signaling, which guide healthy lung development early in life. However, mouse embryos lacking Bmpr1a developed abnormal lung cysts that were similar to those found in CPAM patients, suggesting that problems with BMP signalling might also trigger CPAM in humans. Luo et al. also identified several other genes in the Bmpr1a-deficient mouse lungs that had abnormal patterns of activity. All these genes were known to be controlled by BMP signaling, and to play a role in the development and organisation of lung tissue. This suggests that when these genes are not controlled properly, they could drive formation of CPAM cysts when BMP signaling is compromised. This work is a significant advance in the tools available to study CPAM. Luo et al.'s results also shed new light on the molecular mechanisms underpinning this rare disorder. In the future, Luo et al. hope this knowledge will help us develop better treatments for CPAM, or even help to prevent it altogether.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I , Pulmão , Mesoderma , Camundongos Knockout , Transdução de Sinais , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Camundongos , Pulmão/embriologia , Pulmão/metabolismo , Pulmão/patologia , Mesoderma/embriologia , Mesoderma/metabolismo , Cistos/metabolismo , Cistos/patologia , Cistos/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Pneumopatias/metabolismo , Pneumopatias/patologia , Pneumopatias/genética , Modelos Animais de Doenças
2.
Oral Dis ; 28(4): 1215-1227, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-33759298

RESUMO

OBJECTIVE: In chondrogenesis, BMP signaling was inferred to exhibit regional specificity during Meckel's cartilage morphogenesis. This study aimed to explore the differences in BMP signaling activity between different parts of Meckel's cartilage and the impacts of BMP4 or ALK3 deficiency on the development of Meckel's cartilage during embryogenesis. MATERIALS AND METHODS: The BRE-gal reporter mouse line was utilized to gain an overall picture of canonical BMP signaling activity, as assessed by X-gal staining. Mouse models lacking either Bmp4 or Alk3 in neural crest cells (Wnt1-Cre;Bmp4fl/fl and Wnt1-Cre;Alk3fl/fl ) were generated to explore the morphogenesis of Meckel's cartilage and the mandibular symphysis, as assessed by skeletal staining, histology, and immunostaining. RESULTS: Different parts of Meckel's cartilage exhibited activation of different combinations of BMP signaling pathways. In Wnt1-Cre;Bmp4fl/fl mutants, Sox9+ condensation of the chondrogenic rostral process failed to form, and the V-shaped Runx2+ tissue was split in the median mandibular symphysis. The Wnt1-Cre;Bmp4fl/fl and Wnt1-Cre;Alk3fl/fl mouse models both exhibited truncated Meckel's cartilage, aberrant mandibular intramembranous bone, and tongue muscle abnormalities. CONCLUSIONS: The central hard-tissue loss of both mutant mouse models led to a mandibular symphysis cleft, mimicking the typical sign of the median mandible Tessier 30 cleft in humans.


Assuntos
Proteína Morfogenética Óssea 4 , Receptores de Proteínas Morfogenéticas Ósseas Tipo I , Condrogênese , Mandíbula , Animais , Proteína Morfogenética Óssea 4/deficiência , Proteína Morfogenética Óssea 4/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Cartilagem/metabolismo , Mandíbula/metabolismo , Camundongos , Crista Neural/metabolismo , Transdução de Sinais
4.
eNeuro ; 6(2)2019.
Artigo em Inglês | MEDLINE | ID: mdl-31028086

RESUMO

Blocking inhibitory factors within CNS demyelinating lesions is regarded as a promising strategy to promote remyelination. Bone morphogenetic protein 4 (BMP4) is an inhibitory factor present in demyelinating lesions. Noggin, an endogenous antagonist to BMP, has previously been shown to increase the number of oligodendrocytes and promote remyelination in vivo. However, it remains unclear how BMP4 signaling inhibits remyelination. Here we investigated the downstream signaling pathway that mediates the inhibitory effect that BMP4 exerts upon remyelination through pharmacological and transgenic approaches. Using the cuprizone mouse model of central demyelination, we demonstrate that selectively blocking BMP4 signaling via the pharmacological inhibitor LDN-193189 significantly promotes oligodendroglial differentiation and the extent of remyelination in vivo This was accompanied by the downregulation of transcriptional targets that suppress oligodendrocyte differentiation. Further, selective deletion of BMP receptor type IA (BMPRIA) within primary mouse oligodendrocyte progenitor cells (OPCs) significantly enhanced their differentiation and subsequent myelination in vitro Together, the results of this study identify that BMP4 signals via BMPRIA within OPCs to inhibit oligodendroglial differentiation and their capacity to myelinate axons, and suggest that blocking the BMP4/BMPRIA pathway in OPCs is a promising strategy to promote CNS remyelination.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Doenças Desmielinizantes/metabolismo , Células-Tronco Neurais/metabolismo , Oligodendroglia/metabolismo , Remielinização , Transdução de Sinais , Animais , Proteína Morfogenética Óssea 4/antagonistas & inibidores , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Diferenciação Celular/efeitos dos fármacos , Doenças Desmielinizantes/tratamento farmacológico , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Neurais/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Pirazóis/farmacologia , Pirimidinas/farmacologia , Remielinização/efeitos dos fármacos
5.
Nat Commun ; 9(1): 4831, 2018 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-30446646

RESUMO

Much of the mammalian skeleton originates from a cartilage template eventually replaced by bone via endochondral ossification. Despite much knowledge about growth factors and nuclear proteins in skeletal development, little is understood about the role of metabolic regulation. Here we report that genetic deletion of the glucose transporter Glut1 (Slc2a1), either before or after the onset of chondrogenesis in the limb, severely impairs chondrocyte proliferation and hypertrophy, resulting in dramatic shortening of the limbs. The cartilage defects are reminiscent to those caused by deficiency in Bmp signaling. Importantly, deletion of Bmpr1a in chondrocytes markedly reduces Glut1 levels in vivo, whereas recombinant BMP2 increases Glut1 mRNA and protein levels, boosting glucose metabolism in primary chondrocytes. Biochemical studies identify a Bmp-mTORC1-Hif1a signaling cascade resulting in upregulation of Glut1 in chondrocytes. The results therefore uncover a hitherto unknown connection between Bmp signaling and glucose metabolism in the regulation of cartilage development.


Assuntos
Proteína Morfogenética Óssea 1/genética , Osso e Ossos/metabolismo , Transportador de Glucose Tipo 1/genética , Glucose/metabolismo , Osteogênese/genética , Transdução de Sinais/genética , Animais , Animais Recém-Nascidos , Proteína Morfogenética Óssea 1/deficiência , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 2/farmacologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Osso e Ossos/citologia , Cartilagem/citologia , Cartilagem/crescimento & desenvolvimento , Cartilagem/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrogênese/genética , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Transportador de Glucose Tipo 1/deficiência , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Knockout , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Cultura Primária de Células
6.
Free Radic Biol Med ; 129: 127-137, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30227271

RESUMO

The bone morphogenetic protein (BMP) type I receptors ALK2 and ALK3 are essential for expression of hepcidin, a key iron regulatory hormone. In mice, hepatocyte-specific Alk2 deficiency leads to moderate iron overload with periportal liver iron accumulation, while hepatocyte-specific Alk3 deficiency leads to severe iron overload with centrilobular liver iron accumulation and a more marked reduction of basal hepcidin levels. The objective of this study was to investigate whether the two receptors have additive roles in hepcidin regulation. Iron overload in mice with hepatocyte-specific Alk2 and Alk3 (Alk2/3) deficiency was characterized and compared to hepatocyte-specific Alk3 deficient mice. Co-immunoprecipitation studies were performed to detect the formation of ALK2 and ALK3 homodimer and heterodimer complexes in vitro in the presence and absence of ligands. The iron overload phenotype of hepatocyte-specific Alk2/3-deficient mice was more severe than that of hepatocyte-specific Alk3-deficient mice. In vitro co-immunoprecipitation studies in Huh7 cells showed that ALK3 can homodimerize in absence of BMP2 or BMP6. In contrast, ALK2 did not homodimerize in either the presence or absence of BMP ligands. However, ALK2 did form heterodimers with ALK3 in the presence of BMP2 or BMP6. ALK3-ALK3 and ALK2-ALK3 receptor complexes induced hepcidin expression in Huh7 cells. Our data indicate that: (I) ALK2 and ALK3 have additive functions in vivo, as Alk2/3 deficiency leads to a greater degree of iron overload than Alk3 deficiency; (II) ALK3, but not ALK2, undergoes ligand-independent homodimerization; (III) the formation of ALK2-ALK3 heterodimers is ligand-dependent and (IV) both receptor complexes functionally induce hepcidin expression in vitro.


Assuntos
Receptores de Ativinas Tipo I/genética , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 6/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Hepcidinas/genética , Sobrecarga de Ferro/genética , Ferro/metabolismo , Receptores de Ativinas Tipo I/deficiência , Animais , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 6/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Linhagem Celular Tumoral , Feminino , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepcidinas/metabolismo , Humanos , Sobrecarga de Ferro/metabolismo , Sobrecarga de Ferro/patologia , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Ligação Proteica , Multimerização Proteica , Índice de Gravidade de Doença , Transdução de Sinais
7.
Sci Rep ; 8(1): 8475, 2018 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-29855498

RESUMO

Bone morphogenetic proteins (BMPs) activate the canonical Smad1/5/8 and non-canonical Tak1-MAPK pathways via BMP receptors I and II to regulate skeletal development and bone remodeling. Specific ablation of Bmpr1a in immature osteoblasts, osteoblasts, or osteocytes results in an increase in cancellous bone mass, yet opposite results have been reported regarding the underlying mechanisms. Moreover, the role for BMPRIA-mediated signaling in bone marrow mesenchymal stromal cells (BM-MSCs) has not been explored. Here, we specifically ablated Bmpr1a in BM-MSCs in adult mice to study the function of BMPR1A in bone remodeling and found that the mutant mice showed an increase in cancellous and cortical bone mass, which was accompanied by a decrease in bone formation rate and a greater decrease in bone resorption. Decreased bone formation was associated with a defect in BM-MSC osteogenic differentiation whereas decreased bone resorption was associated with a decrease in RANKL production and osteoclastogenesis. However, ablation of Tak1, a critical non-canonical signaling molecule downstream of BMP receptors, in BM-MSCs at adult stage did not affect bone remodeling. These results suggest that BMP signaling through BMPRIA controls BM-MSC osteogenic differentiation/bone formation and RANKL expression/osteoclastogenesis in adult mice independent of Tak1 signaling.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Diferenciação Celular , Ligante RANK/metabolismo , Animais , Células da Medula Óssea/citologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Linhagem da Célula , Proteínas de Homeodomínio/metabolismo , MAP Quinase Quinase Quinases/deficiência , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Transgênicos , Osteoblastos/citologia , Osteoblastos/metabolismo , Ligante RANK/genética , Transdução de Sinais
8.
BMC Physiol ; 18(1): 3, 2018 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-29482530

RESUMO

BACKGROUND: Inflammatory stimuli induce the hepatic iron regulatory hormone hepcidin, which contributes to anaemia of inflammation (AI). Hepcidin expression is regulated by the bone morphogenetic protein (BMP) and the interleukin-6 (IL-6) signalling pathways. Prior results indicate that the BMP type I receptor ALK3 is mainly involved in the acute inflammatory hepcidin induction four and 72 h after IL-6 administration. In this study, the role of ALK3 in a chronic model of inflammation was investigated. The intact, heat-killed bacterium Brucella abortus (BA) was used to analyse its effect on the development of inflammation and hypoferremia in mice with hepatocyte-specific Alk3-deficiency (Alk3fl/fl; Alb-Cre) compared to control (Alk3fl/fl) mice. RESULTS: An iron restricted diet prevented development of the iron overload phenotype in mice with hepatocyte-specific Alk3 deficiency. Regular diet leads to iron overload and increased haemoglobin levels in these mice, which protects from the development of AI per se. Fourteen days after BA injection Alk3fl/fl; Alb-Cre mice presented milder anaemia (Hb 16.7 g/dl to 11.6 g/dl) compared to Alk3fl/fl control mice (Hb 14.9 g/dl to 8.6 g/dl). BA injection led to an intact inflammatory response in all groups of mice. In Alk3fl/fl; Alb-Cre mice, SMAD1/5/8 phosphorylation was reduced after BA as well as after infection with Staphylococcus aureus. The reduction of the SMAD1/5/8 signalling pathway due to hepatocyte-specific Alk3 deficiency partly suppressed the induction of STAT3 signalling. CONCLUSION: The results reveal in vivo, that 1) hepatocyte-specific Alk3 deficiency partly protects from AI, 2) the development of hypoferremia is partly dependent on ALK3, and 3) the ALK3/BMP/hepcidin axis may serve as a possible therapeutic target to attenuate AI.


Assuntos
Anemia/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Inflamação/metabolismo , Anemia/complicações , Animais , Brucella abortus , Brucelose/complicações , Eritropoese , Feminino , Hepatócitos/metabolismo , Inflamação/etiologia , Mediadores da Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Fosforilação , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo
9.
Mol Ther ; 25(8): 1974-1987, 2017 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-28716575

RESUMO

Trauma-induced heterotopic ossification (tHO) is a condition of pathologic wound healing, defined by the progressive formation of ectopic bone in soft tissue following severe burns or trauma. Because previous studies have shown that genetic variants of HO, such as fibrodysplasia ossificans progressiva (FOP), are caused by hyperactivating mutations of the type I bone morphogenetic protein receptor (T1-BMPR) ACVR1/ALK2, studies evaluating therapies for HO have been directed primarily toward drugs for this specific receptor. However, patients with tHO do not carry known T1-BMPR mutations. Here we show that, although BMP signaling is required for tHO, no single T1-BMPR (ACVR1/ALK2, BMPR1a/ALK3, or BMPR1b/ALK6) alone is necessary for this disease, suggesting that these receptors have functional redundancy in the setting of tHO. By utilizing two different classes of BMP signaling inhibitors, we developed a translational approach to treatment, integrating treatment choice with existing diagnostic options. Our treatment paradigm balances either immediate therapy with reduced risk for adverse effects (Alk3-Fc) or delayed therapy with improved patient selection but greater risk for adverse effects (LDN-212854).


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas/genética , Marcação de Genes , Ossificação Heterotópica/etiologia , Ossificação Heterotópica/patologia , Ferimentos e Lesões/complicações , Receptores de Ativinas Tipo I/deficiência , Animais , Anti-Inflamatórios/farmacologia , Biomarcadores , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Técnicas de Inativação de Genes , Predisposição Genética para Doença , Humanos , Ligantes , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Neutrófilos/metabolismo , Ossificação Heterotópica/prevenção & controle , Inibidores de Proteínas Quinases/farmacologia
10.
Arterioscler Thromb Vasc Biol ; 37(8): 1559-1569, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28619995

RESUMO

OBJECTIVE: We determined in patients with pulmonary arterial (PA) hypertension (PAH) whether in addition to increased production of elastase by PA smooth muscle cells previously reported, PA elastic fibers are susceptible to degradation because of their abnormal assembly. APPROACH AND RESULTS: Fibrillin-1 and elastin are the major components of elastic fibers, and fibrillin-1 binds bone morphogenetic proteins (BMPs) and the large latent complex of transforming growth factor-ß1 (TGFß1). Thus, we considered whether BMPs like TGFß1 contribute to elastic fiber assembly and whether this process is perturbed in PAH particularly when the BMP receptor, BMPR2, is mutant. We also assessed whether in mice with Bmpr2/1a compound heterozygosity, elastic fibers are susceptible to degradation. In PA smooth muscle cells and adventitial fibroblasts, TGFß1 increased elastin mRNA, but the elevation in elastin protein was dependent on BMPR2; TGFß1 and BMP4, via BMPR2, increased extracellular accumulation of fibrillin-1. Both BMP4- and TGFß1-stimulated elastic fiber assembly was impaired in idiopathic (I) PAH-PA adventitial fibroblast versus control cells, particularly those with hereditary (H) PAH and a BMPR2 mutation. This was related to profound reductions in elastin and fibrillin-1 mRNA. Elastin protein was increased in IPAH PA adventitial fibroblast by TGFß1 but only minimally so in BMPR2 mutant cells. Fibrillin-1 protein increased only modestly in IPAH or HPAH PA adventitial fibroblasts stimulated with BMP4 or TGFß1. In Bmpr2/1a heterozygote mice, reduced PA fibrillin-1 was associated with elastic fiber susceptibility to degradation and more severe pulmonary hypertension. CONCLUSIONS: Disrupting BMPR2 impairs TGFß1- and BMP4-mediated elastic fiber assembly and is of pathophysiologic significance in PAH.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Tecido Elástico/metabolismo , Hipertensão Pulmonar Primária Familiar/metabolismo , Hipertensão Pulmonar/metabolismo , Artéria Pulmonar/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Remodelação Vascular , Animais , Proteína Morfogenética Óssea 4/farmacologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Estudos de Casos e Controles , Células Cultivadas , Modelos Animais de Doenças , Tecido Elástico/patologia , Tecido Elástico/fisiopatologia , Elastina/genética , Elastina/metabolismo , Hipertensão Pulmonar Primária Familiar/genética , Hipertensão Pulmonar Primária Familiar/patologia , Hipertensão Pulmonar Primária Familiar/fisiopatologia , Fibrilina-1/genética , Fibrilina-1/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Predisposição Genética para Doença , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/fisiopatologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Fenótipo , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Artéria Pulmonar/fisiopatologia , Interferência de RNA , Transfecção
11.
Arterioscler Thromb Vasc Biol ; 37(4): 657-663, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28232325

RESUMO

OBJECTIVE: Increasing evidence suggests that bone morphogenetic protein (BMP) signaling regulates angiogenesis. Here, we aimed to define the function of BMP receptors in regulating early postnatal angiogenesis by analysis of inducible, endothelial-specific deletion of the BMP receptor components Bmpr2 (BMP type 2 receptor), Alk1 (activin receptor-like kinase 1), Alk2, and Alk3 in mouse retinal vessels. APPROACH AND RESULTS: Expression analysis of several BMP ligands showed that proangiogenic BMP ligands are highly expressed in postnatal retinas. Consistently, BMP receptors are also strongly expressed in retina with a distinct pattern. To assess the function of BMP signaling in retinal angiogenesis, we first generated mice carrying an endothelial-specific inducible deletion of Bmpr2. Postnatal deletion of Bmpr2 in endothelial cells substantially decreased the number of angiogenic sprouts at the vascular front and branch points behind the front, leading to attenuated radial expansion. To identify critical BMPR1s (BMP type 1 receptors) associated with BMPR2 in retinal angiogenesis, we generated endothelial-specific inducible deletion of 3 BMPR1s abundantly expressed in endothelial cells and analyzed the respective phenotypes. Among these, endothelial-specific deletion of either Alk2/acvr1 or Alk3/Bmpr1a caused a delay in radial expansion, reminiscent of vascular defects associated with postnatal endothelial-specific deletion of BMPR2, suggesting that ALK2/ACVR1 and ALK3/BMPR1A are likely to be the critical BMPR1s necessary for proangiogenic BMP signaling in retinal vessels. CONCLUSIONS: Our data identify BMP signaling mediated by coordination of ALK2/ACVR1, ALK3/BMPR1A, and BMPR2 as an essential proangiogenic cue for retinal vessels.


Assuntos
Receptores de Ativinas Tipo I/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Células Endoteliais/efeitos dos fármacos , Artéria Retiniana/efeitos dos fármacos , Neovascularização Retiniana , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Ligantes , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Artéria Retiniana/metabolismo , Transdução de Sinais
12.
Arterioscler Thromb Vasc Biol ; 36(7): 1398-405, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27199449

RESUMO

OBJECTIVE: Calcific aortic valve disease (CAVD) is the most prevalent type of heart valve disease, affecting ≈2% of the US population. CAVD is characterized by the presence of calcific nodules, resulting in aortic valve (AoV) stenosis; however, the underlying mechanisms driving disease remain unknown. Studies of human diseased AoV provide initial evidence that bone morphogenetic protein (BMP) signaling, essential for normal bone formation, is activated during CAVD. Mice deficient in Klotho, an FGF23 transmembrane coreceptor, exhibit premature aging and develop AoV calcific nodules as occurs in human CAVD. The role of BMP signaling in the development of CAVD was examined in porcine aortic valve interstitial cells (VICs) and Klotho(-/-) mice. APPROACH AND RESULTS: We show that activation of BMP signaling, as indicated by pSmad1/5/8 expression, precedes and later localizes with AoV calcification in Klotho(-/-) mice. In addition, cellular and extracellular matrix changes resembling features of normal bone formation are accompanied by increased osteochondrogenic gene induction in calcified Klotho(-/-) AoV. Likewise, osteogenic media treatment of porcine VICs results in BMP pathway activation, increased osteochondrogenic gene induction, and formation of calcific nodules in vitro. We demonstrate that genetic inactivation of the BMP type IA receptor in Klotho(-/-) aortic VICs, as well as BMP pathway inhibition of osteogenic media-treated aortic VICs in vitro, results in the inhibition of AoV calcification. CONCLUSIONS: BMP signaling and osteochondrogenic gene induction are active in calcified Klotho(-/-) AoV in vivo and calcified porcine aortic VICs in vitro. Importantly, BMP signaling is required for the development of AoV calcification in vitro and in vivo.


Assuntos
Estenose da Valva Aórtica/metabolismo , Valva Aórtica/metabolismo , Valva Aórtica/patologia , Proteínas Morfogenéticas Ósseas/metabolismo , Calcinose/metabolismo , Transdução de Sinais , Proteína Smad5/metabolismo , Animais , Valva Aórtica/efeitos dos fármacos , Estenose da Valva Aórtica/genética , Estenose da Valva Aórtica/patologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Calcinose/genética , Calcinose/patologia , Células Cultivadas , Condrogênese , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Feminino , Fator de Crescimento de Fibroblastos 23 , Regulação da Expressão Gênica , Predisposição Genética para Doença , Glucuronidase/deficiência , Glucuronidase/genética , Proteínas Klotho , Masculino , Camundongos Knockout , Osteogênese , Fenótipo , Fosforilação , Pirazóis/farmacologia , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/metabolismo , Proteína Smad8/metabolismo , Suínos
13.
J Endocrinol ; 229(3): 331-41, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27029473

RESUMO

Pituitary follicle-stimulating hormone (FSH) synthesis is regulated by transforming growth factorßsuperfamily ligands, most notably the activins and inhibins. Bone morphogenetic proteins (BMPs) also regulate FSHß subunit (Fshb) expression in immortalized murine gonadotrope-like LßT2 cells and in primary murine or ovine primary pituitary cultures. BMP2 signals preferentially via the BMP type I receptor, BMPR1A, to stimulate murine Fshb transcription in vitro Here, we used a Cre-lox approach to assess BMPR1A's role in FSH synthesis in mice in vivo Gonadotrope-specific Bmpr1a knockout animals developed normally and had reproductive organ weights comparable with those of controls. Knockouts were fertile, with normal serum gonadotropins and pituitary gonadotropin subunit mRNA expression. Cre-mediated recombination of the floxed Bmpr1a allele was efficient and specific, as indicated by PCR analysis of diverse tissues and isolated gonadotrope cells. Furthermore, BMP2 stimulation of inhibitor of DNA binding 3 expression was impaired in gonadotropes isolated from Bmpr1a knockout mice, confirming the loss of functional receptor protein in these cells. Treatment of purified gonadotropes with small-molecule inhibitors of BMPR1A (and the related receptors BMPR1B and ACVR1) suppressed Fshb mRNA expression, suggesting that an autocrine BMP-like molecule might regulate FSH synthesis. However, deletion of Bmpr1a and Acvr1 in cultured pituitary cells did not alter Fshb expression, indicating that the inhibitors had off-target effects. In sum, BMPs or related ligands acting via BMPR1A or ACVR1 are unlikely to play direct physiological roles in FSH synthesis by murine gonadotrope cells.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Fertilidade/fisiologia , Gonadotrofos/fisiologia , Gonadotropinas Hipofisárias/biossíntese , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/fisiologia , Animais , Proteína Morfogenética Óssea 2/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/antagonistas & inibidores , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/fisiologia , Células Cultivadas , Feminino , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Subunidade beta do Hormônio Folículoestimulante/genética , Gonadotrofos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
14.
Am J Physiol Heart Circ Physiol ; 310(8): H984-94, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26873969

RESUMO

Bone morphogenetic protein (BMP) signaling contributes to the development of cardiac hypertrophy. However, the identity of the BMP type I receptor involved in cardiac hypertrophy and the underlying molecular mechanisms are poorly understood. By using quantitative PCR and immunoblotting, we demonstrated that BMP signaling increased during phenylephrine-induced hypertrophy in cultured neonatal rat cardiomyocytes (NRCs), as evidenced by increased phosphorylation of Smads 1 and 5 and induction of Id1 gene expression. Inhibition of BMP signaling with LDN193189 or noggin, and silencing of Smad 1 or 4 using small interfering RNA diminished the ability of phenylephrine to induce hypertrophy in NRCs. Conversely, activation of BMP signaling with BMP2 or BMP4 induced hypertrophy in NRCs. Luciferase reporter assay further showed that BMP2 or BMP4 treatment of NRCs repressed atrogin-1 gene expression concomitant with an increase in calcineurin protein levels and enhanced activity of nuclear factor of activated T cells, providing a mechanism by which BMP signaling contributes to cardiac hypertrophy. In a model of cardiac hypertrophy, C57BL/6 mice treated with angiotensin II (A2) had increased BMP signaling in the left ventricle. Treatment with LDN193189 attenuated A2-induced cardiac hypertrophy and collagen deposition in left ventricles. Cardiomyocyte-specific deletion of BMP type I receptor ALK2 (activin-like kinase 2), but not ALK1 or ALK3, inhibited BMP signaling and mitigated A2-induced cardiac hypertrophy and left ventricular fibrosis in mice. The results suggest that BMP signaling upregulates the calcineurin/nuclear factor of activated T cell pathway via BMP type I receptor ALK2, contributing to cardiac hypertrophy and fibrosis.


Assuntos
Receptores de Ativinas Tipo I/metabolismo , Angiotensina II , Proteína Morfogenética Óssea 2/farmacologia , Proteína Morfogenética Óssea 4/farmacologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Cardiomegalia/enzimologia , Miócitos Cardíacos/enzimologia , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Cardiomegalia/induzido quimicamente , Cardiomegalia/genética , Cardiomegalia/patologia , Cardiomegalia/prevenção & controle , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Fibrose , Proteína 1 Inibidora de Diferenciação/genética , Proteína 1 Inibidora de Diferenciação/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Fatores de Transcrição NFATC/metabolismo , Fenilefrina/farmacologia , Fosforilação , Pirazóis/farmacologia , Pirimidinas/farmacologia , Interferência de RNA , Ratos Sprague-Dawley , Transdução de Sinais , Proteínas Smad/genética , Proteínas Smad/metabolismo , Fatores de Tempo , Transfecção
15.
Proc Natl Acad Sci U S A ; 113(3): E387-95, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26721398

RESUMO

The window of implantation is defined by the inhibition of uterine epithelial proliferation, structural epithelial cell remodeling, and attenuated estrogen (E2) response. These changes occur via paracrine signaling between the uterine epithelium and stroma. Because implantation defects are a major cause of infertility in women, identifying these signaling pathways will improve infertility interventions. Bone morphogenetic proteins (BMPs) are TGF-ß family members that regulate the postimplantation and midgestation stages of pregnancy. In this study, we discovered that signaling via activin-like kinase 3 (ALK3/BMPR1A), a BMP type 1 receptor, is necessary for blastocyst attachment. Conditional knockout (cKO) of ALK3 in the uterus was obtained by producing Alk3(flox) (/flox)-Pgr-cre-positive females. Alk3 cKO mice are sterile and have defects in the luminal uterine epithelium, including increased microvilli density and maintenance of apical cell polarity. Moreover, Alk3 cKO mice exhibit an elevated uterine E2 response and unopposed epithelial cell proliferation during the window of implantation. We determined that dual transcriptional regulation of Kruppel-like factor 15 (Klf15), by both the transforming growth factor ß (TGF-ß) transcription factor SMAD family member 4 (SMAD4) and progesterone receptor (PR), is necessary to inhibit uterine epithelial cell proliferation, a key step for embryo implantation. Our findings present a convergence of BMP and steroid hormone signaling pathways in the regulation of uterine receptivity.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Implantação do Embrião , Útero/metabolismo , Animais , Blastocisto/efeitos dos fármacos , Blastocisto/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Proliferação de Células/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Decídua/efeitos dos fármacos , Decídua/metabolismo , Implantação do Embrião/efeitos dos fármacos , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Estradiol/farmacologia , Feminino , Deleção de Genes , Perfilação da Expressão Gênica , Infertilidade Feminina/metabolismo , Fatores de Transcrição Kruppel-Like , Camundongos Knockout , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Ovário/efeitos dos fármacos , Ovário/fisiologia , Ovário/transplante , Gravidez , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteína Smad4/metabolismo , Fatores de Transcrição/metabolismo , Útero/efeitos dos fármacos , Útero/ultraestrutura
16.
Oncotarget ; 6(26): 22890-904, 2015 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-26274893

RESUMO

Bone Morphogenetic Proteins (BMPs) are secreted cytokines/growth factors belonging to the Transforming Growth Factor ß (TGFß) family. BMP ligands have been shown to be overexpressed in human breast cancers. Normal and cancerous breast tissue display active BMP signaling as indicated by phosphorylated Smads 1, 5 and 9. We combined mice expressing the MMTV.PyMT oncogene with mice having conditional knockout (cKO) of BMP receptor type 1a (BMPR1a) using whey acidic protein (WAP)-Cre and found this deletion resulted in delayed tumor onset and significantly extended survival. Immunofluorescence staining revealed that cKO tumors co-expressed Keratin 5 and mesenchymal cell markers such as Vimentin. This indicates that epithelial-to-mesenchymal (EMT)-like transitions occurred in cKO tumors. We performed microarray analysis on these tumors and found changes that support EMT-like changes. We established primary tumor cell lines and found that BMPR1a cKO had slower growth in vitro and in vivo upon implantation. cKO tumor cells had reduced migration in vitro. We analyzed human databases from TCGA and survival data from microarrays to confirm BMPR1a tumor promoting functions, and found that high BMPR1a gene expression correlates with decreased survival regardless of molecular breast cancer subtype. In conclusion, the data indicate that BMP signaling through BMPR1a functions as a tumor promoter.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Neoplasias da Mama/metabolismo , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Análise em Microsséries , Metástase Neoplásica , Transdução de Sinais
17.
Am J Med Genet A ; 167(6): 1305-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25846706

RESUMO

We describe a patient with a severe juvenile polyposis phenotype, due to a de novo deletion of chromosome 10q22.3-q24.1. He was initially diagnosed with Juvenile polyposis syndrome (JPS) at age four after presenting with hematochezia due to multiple colonic juvenile polyps. He then re-presented at 23 years with recurrent hematochezia from juvenile polyps in his ileoanal pouch. He is one of the earliest reported cases of JPS associated with a large deletion of chromosome 10. Since his initial diagnosis of JPS further studies have confirmed an association between JPS and mutations in BMPR1A in chromosome band 10q23.2, which is in close proximity to PTEN. Mutations in PTEN cause Cowden syndrome (CS) and other PTEN hamartoma tumor syndromes. Due to the chromosome 10 deletion involving contiguous portions of BMPR1A and PTEN in our patient, he may be at risk for CS associated cancers and features, in addition to the polyps associated with JPS. This case presents new challenges in developing appropriate surveillance algorithms to account for the risks associated with each syndrome and highlights the importance of longitudinal follow-up and transitional care between pediatric and adult gastroenterology for patients with hereditary polyposis syndromes.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Deleção Cromossômica , Cromossomos Humanos Par 10 , Hemorragia Gastrointestinal/genética , Síndrome do Hamartoma Múltiplo/genética , Polipose Intestinal/congênito , Síndromes Neoplásicas Hereditárias/genética , PTEN Fosfo-Hidrolase/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Hemorragia Gastrointestinal/patologia , Hemorragia Gastrointestinal/cirurgia , Expressão Gênica , Genótipo , Síndrome do Hamartoma Múltiplo/patologia , Síndrome do Hamartoma Múltiplo/cirurgia , Humanos , Polipose Intestinal/genética , Polipose Intestinal/patologia , Polipose Intestinal/cirurgia , Masculino , Monitorização Fisiológica , Síndromes Neoplásicas Hereditárias/patologia , Síndromes Neoplásicas Hereditárias/cirurgia , PTEN Fosfo-Hidrolase/deficiência , Fenótipo , Adulto Jovem
18.
PLoS One ; 9(12): e113785, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25470749

RESUMO

Platelet-derived growth factors (PDGFs) play important roles in skeletal development and bone fracture healing, yet how PDGFs execute their functions remains incompletely understood. Here we show that PDGF-AA, but not -AB or -BB, could activate the BMP-Smad1/5/8 pathway in mesenchymal stem cells (MSCs), which requires BMPRIA as well as PDGFRα. PDGF-AA promotes MSC osteogenic differentiation through the BMP-Smad1/5/8-Runx2/Osx axis and MSC migration via the BMP-Smad1/5/8-Twist1/Atf4 axis. Mechanistic studies show that PDGF-AA activates BMP-Smad1/5/8 signaling by feedback down-regulating PDGFRα, which frees BMPRI and allows for BMPRI-BMPRII complex formation to activate smad1/5/8, using BMP molecules in the microenvironment. This study unravels a physical and functional interaction between PDGFRα and BMPRI, which plays an important role in MSC differentiation and migration, and establishes a link between PDGF-AA and BMPs pathways, two essential regulators of embryonic development and tissue homeostasis.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Smad/metabolismo , Animais , Western Blotting , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteogênese/genética , Ligação Proteica , Interferência de RNA , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo
19.
PLoS One ; 8(11): e81551, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24312319

RESUMO

Anti-Müllerian hormone (AMH) is a member of the TGF-ß superfamily secreted by the gonads of both sexes. This hormone is primarily known for its role in the regression of the Müllerian ducts in male fetuses. In females, AMH is expressed in granulosa cells of developing follicles. Like other members of the TGF-ß superfamily, AMH transduces its signal through two transmembrane serine/threonine kinase receptors including a well characterized type II receptor, AMHR-II. The complete signalling pathway of AMH involving Smads proteins and the type I receptor is well known in the Müllerian duct and in Sertoli and Leydig cells but not in granulosa cells. In addition, few AMH target genes have been identified in these cells. Finally, while several co-receptors have been reported for members of the TGF-ß superfamily, none have been described for AMH. Here, we have shown that none of the Bone Morphogenetic Proteins (BMPs) co-receptors, Repulsive guidance molecules (RGMs), were essential for AMH signalling. We also demonstrated that the main Smad proteins used by AMH in granulosa cells were Smad 1 and Smad 5. Like for the other AMH target cells, the most important type I receptor for AMH in these cells was BMPR-IA. Finally, we have identified a new AMH target gene, Id3, which could be involved in the effects of AMH on the differentiation of granulosa cells and its other target cells.


Assuntos
Hormônio Antimülleriano/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Animais , Hormônio Antimülleriano/farmacologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Feminino , Fibrinolisina/metabolismo , Técnicas de Inativação de Genes , Células da Granulosa/efeitos dos fármacos , Humanos , Camundongos , Transporte Proteico/efeitos dos fármacos , Proteólise , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo
20.
J Neurosci ; 33(38): 15132-44, 2013 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-24048844

RESUMO

Dendrite development is controlled by the interplay of intrinsic and extrinsic signals affecting initiation, growth, and maintenance of complex dendrites. Bone morphogenetic proteins (BMPs) stimulate dendrite growth in cultures of sympathetic, cortical, and hippocampal neurons but it was unclear whether BMPs control dendrite morphology in vivo. Using a conditional knock-out strategy to eliminate Bmpr1a and Smad4 in immature noradrenergic sympathetic neurons we now show that dendrite length, complexity, and neuron cell body size are reduced in adult mice deficient of Bmpr1a. The combined deletion of Bmpr1a and Bmpr1b causes no further decrease in dendritic features. Sympathetic neurons devoid of Bmpr1a/1b display normal Smad1/5/8 phosphorylation, which suggests that Smad-independent signaling paths are involved in dendritic growth control downstream of BMPR1A/B. Indeed, in the Smad4 conditional knock-out dendrite and cell body size are not affected and dendrite complexity and number are increased. Together, these results demonstrate an in vivo function for BMPs in the generation of mature sympathetic neuron dendrites. BMPR1 signaling controls dendrite complexity postnatally during the major dendritic growth period of sympathetic neurons.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Dendritos/metabolismo , Gânglios Simpáticos/citologia , Células Receptoras Sensoriais/citologia , Transdução de Sinais/fisiologia , Fatores Etários , Análise de Variância , Animais , Animais Recém-Nascidos , Proteínas Arqueais/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Proteínas Morfogenéticas Ósseas/genética , Células Cultivadas , DNA Polimerase Dirigida por DNA/metabolismo , Embrião de Mamíferos , Corantes Fluorescentes/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Homeodomínio/metabolismo , Imageamento Tridimensional , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Neurológicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteína Smad4/deficiência , Proteína Smad4/genética , Estatísticas não Paramétricas , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...