Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Clin Chest Med ; 42(1): 217-228, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33541615

RESUMO

Pulmonary arterial hypertension (PAH) occurs in women more than men whereas survival in men is worse than in women. In recent years, much research has been carried out to understand these sex differences in PAH. This article discusses clinical and preclinical studies that have investigated the influences of sex, serotonin, obesity, estrogen, estrogen synthesis, and estrogen metabolism on bone morphogenetic protein receptor type II signaling, the pulmonary circulation and right ventricle in both heritable and idiopathic pulmonary hypertension.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Ventrículos do Coração/fisiopatologia , Hipertensão Pulmonar/fisiopatologia , Circulação Pulmonar/fisiologia , Estrogênios/biossíntese , Estrogênios/metabolismo , Feminino , Humanos , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/genética , Masculino , Obesidade/complicações , Serotonina/metabolismo , Caracteres Sexuais , Transdução de Sinais
2.
Sci Signal ; 14(667)2021 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-33500333

RESUMO

Pulmonary arterial hypertension (PAH) is a fatal disease characterized by excessive pulmonary vascular remodeling. However, despite advances in therapeutic strategies, patients with PAH bearing mutations in the bone morphogenetic protein receptor type 2 (BMPR2)-encoding gene present severe phenotypes and outcomes. We sought to investigate the effect of PER-like kinase (PERK), which participates in one of three major pathways associated with the unfolded protein response (UPR), on PAH pathophysiology in BMPR2 heterozygous mice. BMPR2 heterozygosity in pulmonary artery smooth muscle cells (PASMCs) decreased the abundance of the antiapoptotic microRNA miR124-3p through the arm of the UPR mediated by PERK. Hypoxia promoted the accumulation of unfolded proteins in BMPR2 heterozygous PASMCs, resulting in increased PERK signaling, cell viability, cellular proliferation, and glycolysis. Proteomic analyses revealed that PERK ablation suppressed PDGFRß-STAT1 signaling and glycolysis in hypoxic BMPR2 heterozygous PASMCs. Furthermore, PERK ablation or PERK inhibition ameliorated pulmonary vascular remodeling in the Sugen/chronic hypoxia model of PAH, irrespective of BMPR2 status. Hence, these findings suggest that PERK inhibition is a promising therapeutic strategy for patients with PAH with or without BMPR2 mutation.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Miócitos de Músculo Liso , Hipertensão Arterial Pulmonar/metabolismo , Artéria Pulmonar , eIF-2 Quinase/fisiologia , Animais , Hipóxia Celular , Sobrevivência Celular , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia
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.
Curr Opin Nephrol Hypertens ; 29(2): 171-179, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31815758

RESUMO

PURPOSE OF REVIEW: Pulmonary arterial hypertension (PAH) is characterized by pulmonary arterial endothelial cell (PAEC) dysfunction and apoptosis, pulmonary arterial smooth muscle cell (PASMC) proliferation, inflammation, vasoconstriction, and metabolic disturbances that include disrupted bone morphogenetic protein receptor (BMPR2)-peroxisome proliferator-activated receptor gamma (PPARγ) axis and DNA damage. Activation of PPARγ improves many of these mechanisms, although erroneous reports on potential adverse effects of thiazolidinedione (TZD)-class PPARγ agonists reduced their clinical use in the past decade. Here, we review recent findings in heart, lung, and kidney research related to the pathobiology of vascular remodeling and tissue fibrosis, and also potential therapeutic effects of the PPARγ agonist pioglitazone. RECENT FINDINGS: Independent of its metabolic effects (improved insulin sensitivity and fatty acid handling), PPARγ activation rescues BMPR2 dysfunction, inhibits TGFß/Smad3/CTGF and TGFß/pSTAT3/pFoxO1 pathways, and induces the PPARγ/apoE axis, inhibiting vascular remodeling. PPARγ activation dampens mtDNA damage via PPARγ/UBR5/ATM pathway, improves function of endothelial progenitor cells (EPCs), and decrease renal fibrosis by repressing TGFß/pSTAT3 and TGFß/EGR1. SUMMARY: Pharmacological PPARγ activation improves many hallmarks of PAH, including dysfunction of BMPR2-PPARγ axis, PAEC, PASMC, EPC, mitochondria/metabolism, and inflammation. Recent randomized controlled trials, including IRIS (Insulin Resistance Intervention After Stroke Trial), emphasize the beneficial effects of PPARγ agonists in PAH patients, leading to recent revival for clinical use.


Assuntos
Matriz Extracelular/fisiologia , Hipertensão Pulmonar/etiologia , Rim/patologia , Músculo Liso Vascular/citologia , Miocárdio/patologia , Miócitos de Músculo Liso/fisiologia , PPAR gama/fisiologia , Fibrose Pulmonar/etiologia , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Dano ao DNA , Fibrose , Homeostase , Humanos , PPAR gama/agonistas , Fator de Crescimento Transformador beta1/fisiologia
5.
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
6.
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
7.
PLoS Biol ; 17(12): e3000557, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31826007

RESUMO

Balanced transforming growth factor-beta (TGFß)/bone morphogenetic protein (BMP)-signaling is essential for tissue formation and homeostasis. While gain in TGFß signaling is often found in diseases, the underlying cellular mechanisms remain poorly defined. Here we show that the receptor BMP type 2 (BMPR2) serves as a central gatekeeper of this balance, highlighted by its deregulation in diseases such as pulmonary arterial hypertension (PAH). We show that BMPR2 deficiency in endothelial cells (ECs) does not abolish pan-BMP-SMAD1/5 responses but instead favors the formation of mixed-heteromeric receptor complexes comprising BMPR1/TGFßR1/TGFßR2 that enable enhanced cellular responses toward TGFß. These include canonical TGFß-SMAD2/3 and lateral TGFß-SMAD1/5 signaling as well as formation of mixed SMAD complexes. Moreover, BMPR2-deficient cells express genes indicative of altered biophysical properties, including up-regulation of extracellular matrix (ECM) proteins such as fibrillin-1 (FBN1) and of integrins. As such, we identified accumulation of ectopic FBN1 fibers remodeled with fibronectin (FN) in junctions of BMPR2-deficient ECs. Ectopic FBN1 deposits were also found in proximity to contractile intimal cells in pulmonary artery lesions of BMPR2-deficient heritable PAH (HPAH) patients. In BMPR2-deficient cells, we show that ectopic FBN1 is accompanied by active ß1-integrin highly abundant in integrin-linked kinase (ILK) mechano-complexes at cell junctions. Increased integrin-dependent adhesion, spreading, and actomyosin-dependent contractility facilitates the retrieval of active TGFß from its latent fibrillin-bound depots. We propose that loss of BMPR2 favors endothelial-to-mesenchymal transition (EndMT) allowing cells of myo-fibroblastic character to create a vicious feed-forward process leading to hyperactivated TGFß signaling. In summary, our findings highlight a crucial role for BMPR2 as a gatekeeper of endothelial homeostasis protecting cells from increased TGFß responses and integrin-mediated mechano-transduction.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Células Endoteliais/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Linhagem Celular , Endotélio Vascular/metabolismo , Fibrilina-1/metabolismo , Regulação da Expressão Gênica/genética , Humanos , Pulmão/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Hipertensão Arterial Pulmonar/metabolismo , Hipertensão Arterial Pulmonar/fisiopatologia , Artéria Pulmonar/metabolismo , Receptores de Fatores de Crescimento Transformadores beta , Transdução de Sinais , Proteínas Smad
8.
J Pathol ; 247(3): 333-346, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30430573

RESUMO

Endothelial-to-mesenchymal transition (EndMT) has been unveiled as a common cause for a multitude of human pathologies, including cancer and cardiovascular disease. Vascular calcification is a risk factor for ischemic vascular disorders and slowing calcification may reduce mortality in affected patients. The absence of early biomarkers hampers the identification of patients at risk. EndMT and vascular calcification are induced upon cooperation between distinct stimuli, including inflammatory cytokines and transforming growth factor beta (TGF-ß) family members. However, how these signaling pathways interplay to promote cell differentiation and eventually vascular calcification is not well understood. Using in vitro and ex vivo analysis in animal models and patient-derived tissues, we have identified that the pro-inflammatory cytokines tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1ß) induce EndMT in human primary aortic endothelial cells, thereby sensitizing them for BMP-9-induced osteogenic differentiation. Downregulation of the BMP type II receptor BMPR2 is a key event in this process. Rather than compromising BMP canonical signal transduction, loss of BMPR2 results in decreased JNK signaling in ECs, thus enhancing BMP-9-induced mineralization. Altogether, our results point at the BMPR2-JNK signaling axis as a key pathway regulating inflammation-induced EndMT and contributing to calcification. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Calcificação Vascular/fisiopatologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Células Endoteliais/fisiologia , Endotélio Vascular/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Humanos , Mediadores da Inflamação/farmacologia , Interleucina-1beta/farmacologia , Camundongos Endogâmicos C3H , Osteogênese/efeitos dos fármacos , Osteogênese/fisiologia , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Calcificação Vascular/patologia
9.
Bone ; 111: 101-108, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29551750

RESUMO

Various substitution mutations in ALK2, a transmembrane serine/threonine kinase receptor for bone morphogenetic proteins (BMPs), have been identified in patients with genetic disorders such as fibrodysplasia ossificans progressiva (FOP), diffuse intrinsic pontine glioma (DIPG) and heart defects. In this study, we characterized the ALK2 mutants R258G, G328V and F246Y, which were identified in patients with severe FOP, DIPG and unusual hereditary skeletal dysplasia, respectively. Both R258G and G328V were gain-of-function mutations, but F246Y was equivalent to wild-type ALK2. We also examined the effect of the suppressor FKBP12 on the signal transduction of a further 14 ALK2 mutations associated with FOP and/or DIPG. To varying extents FKBP12 over-expression suppressed the basal signaling induced by thirteen of the ALK2 mutants, whereas PF197-8L was uniquely resistant. In the PF197-8L mutant, the modelled ALK2 residue L197 induced a steric clash with the D36 residue in FKBP12 and dissociated their interaction. The co-expression of BMP type II receptors or stimulation with ligands relieved the suppression by FKBP12 by disrupting the interaction between mutant ALK2 and FKBP12. Taken together, FKBP12 binds to and suppresses mutant ALK2 proteins associated with FOP and DIPG, except for PF197-8L.


Assuntos
Receptores de Ativinas Tipo I/genética , Doenças do Desenvolvimento Ósseo/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Neoplasias do Tronco Encefálico/genética , Glioma/genética , Miosite Ossificante/genética , Proteína 1A de Ligação a Tacrolimo/fisiologia , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Linhagem Celular , Humanos , Camundongos , Miosite Ossificante/patologia , Ossificação Heterotópica/genética , Transdução de Sinais
10.
J Mol Med (Berl) ; 96(5): 427-444, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29523914

RESUMO

We report the role of miR-1187 in regulation of osteoblast functions. Over-expression of miR-1187 inhibited osteoblast differentiation. Target prediction analysis tools and experimental validation by luciferase 3' UTR reporter assay identified BMPR-II and ArhGEF-9 as direct targets of miR-1187. ArhGEF-9 activates Cdc42 which has a major role in actin reorganization. BMP-2 also induces actin polymerization. Role of miR-1187 in actin reorganization was determined by western blotting, immunofluorescence, and in vivo gene silencing studies. Reduced protein levels of BMPR-II, activated Cdc42, and downstream signaling molecules were observed in miR-1187-transfected osteoblasts. miR-1187 over-expression resulted in decreased actin polymerization. Additionally, P-cofilin, which does not bind F-actin, was decreased in miR-1187-transfected cells. These results were corroborated by administration of BMPR-II exogenously in miR-1187-transfected osteoblasts. Silencing of miR-1187 in neonatal mice mitigated all the inhibitory effects of miR-1187 on actin cytoskeletal rearrangement. Importantly, in vivo treatment of miR-1187 inhibitor to ovariectomized BALB/c mice led to significant improvement in trabecular bone microarchitecture. Overall, miR-1187 functions as a negative regulator of osteogenesis by repressing BMPR-II and ArhGEF-9 expression thus suppressing non-Smad BMP2/Cdc42 signaling pathway and inhibiting actin reorganization. miR-1187 functions as a negative regulator of osteogenesis by repressing BMPR-II expression, which in turn, suppresses non-Smad BMP2/Cdc42 signaling pathway, thus inhibiting actin cytoskeletal rearrangement. Silencing of miR-1187 significantly improves trabecular bone microarchitecture. As miR-1187 exerts a negative regulatory role in osteoblasts function, hence, we propose that therapeutic approaches targeting miR-1187 could be useful in enhancing the bone formation and treatment of pathological conditions of bone loss.


Assuntos
Citoesqueleto de Actina/fisiologia , MicroRNAs/fisiologia , Osteoblastos/fisiologia , Actinas/fisiologia , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Diferenciação Celular , Feminino , Camundongos Endogâmicos BALB C , Fatores de Troca de Nucleotídeo Guanina Rho/fisiologia
11.
Biol Aujourdhui ; 210(2): 65-78, 2016.
Artigo em Francês | MEDLINE | ID: mdl-27687598

RESUMO

Pulmonary arterial hypertension (PAH) is a disorder in which mechanical obstruction of the pulmonary vascular bed is largely responsible for the rise in mean pulmonary arterial pressure (mPAP), resulting in a progressive functional decline despite current available therapeutic options. There are multiple mechanisms predisposing to and/or promoting the aberrant pulmonary vascular remodeling in PAH, and these involve not only altered crosstalk between cells within the vascular wall but also sustained inflammation and dysimmunity, cell accumulation in the vascular wall and excessive activation of some growth factor-stimulated signaling pathways, in addition to the interaction of systemic hormones, local growth factors, cytokines, and transcription factors. Heterozygous germline mutations in the bone morphogenetic protein receptor, type-2 (BMPR2) gene, a gene encoding a receptor for the transforming growth factor (TGF)-ß superfamily, can predispose to the disease. Although the spectrum of therapeutic options for PAH has expanded in the last 20 years, available therapies remain essentially palliative. Over the past decade, however, a better understanding of key regulators of this irreversible remodeling of the pulmonary vasculature has been obtained. New and more effective approaches are likely to emerge. The present article profiles the innovative research into novel pathways and therapeutic targets that may lead to the development of targeted agents in PAH.


Assuntos
Comunicação Celular/fisiologia , Hipertensão Pulmonar/terapia , Terapia de Alvo Molecular/tendências , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Humanos , Hipertensão Pulmonar/imunologia , Hipertensão Pulmonar/metabolismo , Terapia de Alvo Molecular/métodos , Proteínas do Tecido Nervoso/fisiologia , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Transdução de Sinais/fisiologia
12.
Biol Aujourdhui ; 210(2): 79-88, 2016.
Artigo em Francês | MEDLINE | ID: mdl-27687599

RESUMO

Serotonin and bone-marrow-derived stem cells participate together in triggering pulmonary hypertension. Our work has shown that the absence of 5-HT2B receptors generates permanent changes in the composition of the blood and bone-marrow in the myeloid lineages, particularly in endothelial cell progenitors. The initial functions of 5-HT2B receptors in pulmonary arterial hypertension (PAH) are restricted to bone-marrow cells. They contribute to the differentiation/proliferation/mobilization of endothelial progenitor cells from the bone-marrow. Those bone-marrow-derived cells have a critical role in the development of pulmonary hypertension and pulmonary vascular remodeling. These data indicate that bone-marrow derived endothelial progenitors play a key role in the pathogenesis of PAH and suggest that interactions involving serotonin and bone morphogenic protein type 2 receptor (BMPR2) could take place at the level of the bone-marrow.


Assuntos
Medula Óssea/fisiologia , Células Endoteliais/fisiologia , Hipertensão Pulmonar/etiologia , Serotonina/fisiologia , Células-Tronco/fisiologia , Animais , Células da Medula Óssea/fisiologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Humanos , Camundongos
13.
Am J Respir Crit Care Med ; 194(6): 719-28, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27077479

RESUMO

RATIONALE: In heritable pulmonary arterial hypertension with germline mutation in the bone morphogenetic protein receptor type 2 (BMPR2) gene, right ventricle (RV) dysfunction is associated with RV lipotoxicity; however, the underlying mechanism for lipid accumulation is not known. OBJECTIVES: We hypothesized that lipid accumulation in cardiomyocytes with BMPR2 mutation occurs owing to alterations in lipid transport and impaired fatty acid oxidation (FAO), which is exacerbated by a high-lipid (Western) diet (WD). METHODS: We used a transgenic mouse model of pulmonary arterial hypertension with mutant BMPR2 and generated a cardiomyocyte cell line with BMPR2 mutation. Electron microscopy and metabolomic analysis were performed on mouse RVs. MEASUREMENTS AND MAIN RESULTS: By metabolomics analysis, we found an increase in long-chain fatty acids in BMPR2 mutant mouse RVs compared with controls, which correlated with cardiac index. BMPR2-mutant cardiomyocytes had increased lipid compared with controls. Direct measurement of FAO in the WD-fed BMPR2-mutant RV showed impaired palmitate-linked oxygen consumption, and metabolomics analysis showed reduced indices of FAO. Using both mutant BMPR2 mouse RVs and cardiomyocytes, we found an increase in the uptake of (14)C-palmitate and fatty acid transporter CD36 that was further exacerbated by WD. CONCLUSIONS: Taken together, our data suggest that impaired FAO and increased expression of the lipid transporter CD36 are key mechanisms underlying lipid deposition in the BMPR2-mutant RV, which are exacerbated in the presence of dietary lipids. These findings suggest important features leading to RV lipotoxicity in pulmonary arterial hypertension and may point to novel areas of therapeutic intervention.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Ventrículos do Coração/química , Lipídeos/análise , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Linhagem Celular , Modelos Animais de Doenças , Ácidos Graxos/metabolismo , Ventrículos do Coração/metabolismo , Ventrículos do Coração/ultraestrutura , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Metabolismo dos Lipídeos/genética , Metabolômica , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Miócitos Cardíacos/metabolismo
14.
PLoS One ; 10(10): e0139860, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26444546

RESUMO

To investigate the role of Bone Morphogenic Protein Receptor Type II (BMPRII) in learning, memory, and exploratory behavior in mice, a tissue-specific knockout of BMPRII in the post-natal hippocampus and forebrain was generated. We found that BMPRII mutant mice had normal spatial learning and memory in the Morris water maze, but showed significantly reduced swimming speeds with increased floating behavior. Further analysis using the Porsolt Swim Test to investigate behavioral despair did not reveal any differences in immobility between mutants and controls. In the Elevated Plus Maze, BMPRII mutants and Smad4 mutants showed reduced anxiety, while in exploratory tests, BMPRII mutants showed more interest in object exploration. These results suggest that loss of BMPRII in the mouse hippocampus and forebrain does not disrupt spatial learning and memory encoding, but instead impacts exploratory and anxiety-related behaviors.


Assuntos
Ansiedade/fisiopatologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Comportamento Exploratório/fisiologia , Prosencéfalo/fisiologia , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/análise , Feminino , Hipocampo/química , Hipocampo/fisiologia , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prosencéfalo/química , Proteína Smad4/fisiologia
15.
Exp Biol Med (Maywood) ; 240(12): 1580-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25854878

RESUMO

Vascular remodeling due to excessive proliferation of endothelial and smooth muscle cells is a hallmark feature of pulmonary hypertension. microRNAs (miRNAs) are a class of small, non-coding RNA fragments that have recently been associated with remodeling of pulmonary arteries, in particular by silencing the bone morphogenetic protein receptor type II (BMPR2). Here we identified a novel pathway involving the concerted action of miR-125a, BMPR2 and cyclin-dependent kinase inhibitors (CDKN) that controls a proliferative phenotype of endothelial cells. An in silico approach predicted miR-125a to target BMPR2. Functional inhibition of miR-125a resulted in increased proliferation of these cells, an effect that was found accompanied by upregulation of BMPR2 and reduced expression of the tumor suppressors CDKN1A (p21) and CDKN2A (p16). These data were confirmed in experimental pulmonary hypertension in vivo. Levels of miR-125a were elevated in lung tissue of hypoxic animals that develop pulmonary hypertension. In contrast, circulating levels of miR-125a were found to be lower in mice with pulmonary hypertension as compared to control mice. Similar findings were observed in a small cohort of patients with precapillary pulmonary hypertension. These translational data emphasize the pathogenetic role of miR-125a in pulmonary vascular remodeling.


Assuntos
Hipertensão Pulmonar/fisiopatologia , MicroRNAs/fisiologia , Remodelação Vascular/fisiologia , Animais , Western Blotting , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Quinases Ciclina-Dependentes/fisiologia , Endotélio Vascular/fisiologia , Feminino , Humanos , Masculino , Camundongos , Reação em Cadeia da Polimerase em Tempo Real
17.
Chest ; 147(2): 529-537, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25644906

RESUMO

Pulmonary arterial hypertension (PAH) is a disorder in which mechanical obstruction of the pulmonary vascular bed is largely responsible for the rise in mean pulmonary arterial pressure, resulting in a progressive functional decline despite current available therapeutic options. The fundamental pathogenetic mechanisms underlying this disorder include pulmonary vasoconstriction, in situ thrombosis, medial hypertrophy, and intimal proliferation, leading to occlusion of the small to mid-sized pulmonary arterioles and the formation of plexiform lesions. Several predisposing or promoting mechanisms that contribute to excessive pulmonary vascular remodeling in PAH have emerged, such as altered crosstalk between cells within the vascular wall, sustained inflammation and dysimmunity, inhibition of cell death, and excessive activation of signaling pathways, in addition to the impact of systemic hormones, local growth factors, cytokines, transcription factors, and germline mutations. Although the spectrum of therapeutic options for PAH has expanded in the last 20 years, available therapies remain essentially palliative. However, over the past decade, a better understanding of new key regulators of this irreversible pulmonary vascular remodeling has been obtained. This review examines the state-of-the-art potential new targets for innovative research in PAH, focusing on (1) the crosstalk between cells within the pulmonary vascular wall, with particular attention to the role played by dysfunctional endothelial cells; (2) aberrant inflammatory and immune responses; (3) the abnormal extracellular matrix function; and (4) altered BMPRII/KCNK3 signaling systems. A better understanding of novel pathways and therapeutic targets will help in the designing of new and more effective approaches for PAH treatment.


Assuntos
Endotélio Vascular/fisiopatologia , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/fisiopatologia , Remodelação Vascular , Coagulação Sanguínea/fisiologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Progressão da Doença , Humanos , MicroRNAs/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Canais de Potássio de Domínios Poros em Tandem/genética , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Pressão Propulsora Pulmonar , Vasodilatação/fisiologia
18.
Am J Respir Crit Care Med ; 191(6): 693-703, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25608111

RESUMO

RATIONALE: Major pulmonary arterial hypertension (PAH) registries report a greater incidence of PAH in women; mutations in the bone morphogenic protein type II receptor (BMPR-II) occur in approximately 80% of patients with heritable PAH (hPAH). OBJECTIVES: We addressed the hypothesis that women may be predisposed to PAH due to normally reduced basal BMPR-II signaling in human pulmonary artery smooth muscle cells (hPASMCs). METHODS: We examined the BMPR-II signaling pathway in hPASMCs derived from men and women with no underlying cardiovascular disease (non-PAH hPASMCs). We also determined the development of pulmonary hypertension in male and female mice deficient in Smad1. MEASUREMENTS AND MAIN RESULTS: Platelet-derived growth factor, estrogen, and serotonin induced proliferation only in non-PAH female hPASMCs. Female non-PAH hPASMCs exhibited reduced messenger RNA and protein expression of BMPR-II, the signaling intermediary Smad1, and the downstream genes, inhibitors of DNA binding proteins, Id1 and Id3. Induction of phospho-Smad1/5/8 and Id protein by BMP4 was also reduced in female hPASMCs. BMP4 induced proliferation in female, but not male, hPASMCs. However, small interfering RNA silencing of Smad1 invoked proliferative responses to BMP4 in male hPASMCs. In male hPASMCs, estrogen decreased messenger RNA and protein expression of Id genes. The estrogen metabolite 4-hydroxyestradiol decreased phospho-Smad1/5/8 and Id expression in female hPASMCs while increasing these in males commensurate with a decreased proliferative effect in male hPASMCs. Female Smad1(+/-) mice developed pulmonary hypertension (reversed by ovariectomy). CONCLUSIONS: We conclude that estrogen-driven suppression of BMPR-II signaling in non-PAH hPASMCs derived from women contributes to a pro-proliferative phenotype in hPASMCs that may predispose women to PAH.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Músculo Liso Vascular/citologia , Artéria Pulmonar/citologia , Animais , Regulação para Baixo , Estrogênios/metabolismo , Estrogênios/fisiologia , Feminino , Humanos , Hipertensão Pulmonar , Masculino , Camundongos , Fatores Sexuais , Transdução de Sinais/fisiologia
19.
Am J Respir Crit Care Med ; 191(6): 665-77, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25606692

RESUMO

RATIONALE: Up to 10% of patients with systemic sclerosis (SSc) develop pulmonary arterial hypertension (PAH). This risk persists throughout the disease and is time dependent, suggesting that SSc is a susceptibility factor. Outcome for SSc-PAH is poor compared with heritable or idiopathic forms, despite clinical and pathological similarities. Although susceptibility in heritable PAH and idiopathic PAH is strongly associated with gene mutations leading to reduced expression of bone morphogenetic protein receptor (BMPR) II, these mutations have not been observed in SSc-PAH. OBJECTIVES: To explore BMPRII expression and function in a mouse model of SSc (TßRIIΔk-fib) that is susceptible to developing pulmonary hypertension and in SSc lung. METHODS: BMPRII and downstream signaling pathways were profiled in lung tissue and fibroblasts from the TßRIIΔk-fib model, which develops pulmonary vasculopathy with pulmonary hypertension that is exacerbated by SU5416. Complementary studies examined SSc or control lung tissue and fibroblasts. MEASUREMENTS AND MAIN RESULTS: Our study shows reduced BMPRII, impaired signaling, and altered receptor turnover activity in a transforming growth factor (TGF)-ß-dependent mouse model of SSc-PAH. Similarly, a significant reduction in BMPRII expression is observed in SSc lung tissue and fibroblasts. Increased proteasomal degradation of BMPRII appears to underlie this and may result from heightened TGF-ß activity. CONCLUSIONS: We found reduced BMPRII protein in patients with SSc-PAH and a relevant mouse model associated with increased proteasomal degradation of BMPRII. Collectively, these results suggest that impaired BMP signaling, resulting from TGF-ß-dependent increased receptor degradation, may promote PAH susceptibility in SSc and provide a unifying mechanism across different forms of PAH.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Hipertensão Pulmonar/etiologia , Escleroderma Sistêmico/complicações , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Western Blotting , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/análise , Modelos Animais de Doenças , Fibroblastos/fisiologia , Humanos , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Pulmão/química , Pulmão/patologia , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase , Complexo de Endopeptidases do Proteassoma/fisiologia , Escleroderma Sistêmico/metabolismo , Fator de Crescimento Transformador beta/análise
20.
Stem Cells Dev ; 24(3): 354-71, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25209090

RESUMO

Body axes and germ layers evolve at gastrulation, and in mammals are driven by many genes; however, what orchestrates the genetic pathways during gastrulation remains elusive. Previously, we presented evidence that microRNA-17 (miRNA-17) family members, miR-17-5p, miR-20a, miR-93, and miR-106a were differentially expressed in mouse embryos and functioned to control differentiation of the stem cell population. Here, we identify function(s) that these miRNAs have during gastrulation. Fluorescent in situ hybridization miRNA probes reveal that these miRNAs are localized at the mid/posterior primitive streak (ps) in distinct populations of primitive ectoderm, mesendoderm, and mesoderm. Seven different miRNA prediction algorithms are identified in silico bone morphogenic protein receptor 2 (Bmpr2) as a target of these miRNAs. Bmpr2 is a member of the TGFß pathway and invokes stage-specific changes during gastrulation. Recently, Bmpr2 was shown regulating cytoskeletal dynamics, cell movement, and invasion. Our previous and current data led to a hypothesis by which members of the miR-17 family influence gastrulation by suppressing Bmpr2 expression at the primitive streak. This suppression influences fate decisions of cells by affecting genes downstream of BMPR2 as well as mesoderm invasion through regulation of actin dynamics.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/fisiologia , Células-Tronco Embrionárias/citologia , Gastrulação/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/fisiologia , Regiões 3' não Traduzidas , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/biossíntese , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem da Célula/genética , Corpos Embrioides , Desenvolvimento Embrionário , Endoderma/metabolismo , Feminino , Hibridização in Situ Fluorescente , Quinases Lim/fisiologia , Masculino , Camundongos , MicroRNAs/antagonistas & inibidores , Simulação de Acoplamento Molecular , Linha Primitiva/metabolismo , Transdução de Sinais , Proteínas Smad/fisiologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA