Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 297(6): 101410, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34785214

RESUMO

Pluripotent stem cells are known to shift their mitochondrial metabolism upon differentiation, but the mechanisms underlying such metabolic rewiring are not fully understood. We hypothesized that during differentiation of human induced pluripotent stem cells (hiPSCs), mitochondria undergo mitophagy and are then replenished by the biogenesis of new mitochondria adapted to the metabolic needs of the differentiated cell. To evaluate mitophagy during iPSC differentiation, we performed live cell imaging of mitochondria and lysosomes in hiPSCs differentiating into vascular endothelial cells using confocal microscopy. We observed a burst of mitophagy during the initial phases of hiPSC differentiation into the endothelial lineage, followed by subsequent mitochondrial biogenesis as assessed by the mitochondrial biogenesis biosensor MitoTimer. Furthermore, hiPSCs undergoing differentiation showed greater mitochondrial oxidation of fatty acids and an increase in ATP levels as assessed by an ATP biosensor. We also found that during mitophagy, the mitochondrial phosphatase PGAM5 is cleaved in hiPSC-derived endothelial progenitor cells and in turn activates ß-catenin-mediated transcription of the transcriptional coactivator PGC-1α, which upregulates mitochondrial biogenesis. These data suggest that mitophagy itself initiates the increase in mitochondrial biogenesis and oxidative metabolism through transcriptional changes during endothelial cell differentiation. In summary, these findings reveal a mitophagy-mediated mechanism for metabolic rewiring and maturation of differentiating cells via the ß-catenin signaling pathway. We propose that such mitochondrial-nuclear cross talk during hiPSC differentiation could be leveraged to enhance the metabolic maturation of differentiated cells.


Assuntos
Reprogramação Celular , Células Endoteliais , Células-Tronco Pluripotentes Induzidas/metabolismo , Mitofagia , Humanos , Proteínas Mitocondriais/metabolismo , Fosforilação Oxidativa , Fosfoproteínas Fosfatases/metabolismo , Transcrição Gênica , beta Catenina/metabolismo
2.
Am J Physiol Cell Physiol ; 318(5): C913-C930, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32159364

RESUMO

Whole exome sequencing (WES) was used in the research of familial pulmonary arterial hypertension (FPAH). CAV1 and KCNK3 were found as two novel candidate genes of FPAH. However, few pathogenic genes were identified in idiopathic pulmonary arterial hypertension (IPAH). We conducted WES in 20 unrelated IPAH patients who did not carry the known PAH-pathogenic variants among BMPR2, CAV1, KCNK3, SMAD9, ALK1, and ENG. We found a total of 4,950 variants in 3,534 genes, including 4,444 single-nucleotide polymorphisms and 506 insertions/deletions (InDels). Through the comprehensive and multilevel analysis, we disclosed several novel signaling cascades significantly connected to IPAH, including variants related to cadherin signaling pathway, dilated cardiomyopathy, glucose metabolism, immune response, mucin-type O-glycosylation, phospholipase C (PLC)-activating G protein-coupled receptor (GPCR) signaling pathway, vascular contraction and generation, and voltage-dependent Ca2+ channels. We also conducted validation studies in five mutant genes related to PLC-activating GPCR signaling pathway potentially involved in intracellular calcium regulation through Sanger sequencing for mutation accuracy, qRT-PCR for mRNA stability, immunofluorescence for subcellular localization, Western blotting for protein level, Fura-2 imaging for intracellular calcium, and proliferation analysis for cell function. The validation experiments showed that those variants in CCR5 and C3AR1 significantly increased the rise of intracellular calcium and the variant in CCR5 profoundly enhanced proliferative capacity of human pulmonary artery smooth muscle cells. Thus, our study suggests that multiple genetically affected signaling pathways take effect together to cause the formation of IPAH and the development of right heart failure and may further provide new therapy targets or putative clues for the present treatments such as limited therapeutic effectiveness of Ca2+ channel blockers.


Assuntos
Hipertensão Pulmonar Primária Familiar/genética , Insuficiência Cardíaca/genética , Receptores CCR5/genética , Receptores de Complemento/genética , Adulto , Bloqueadores dos Canais de Cálcio/efeitos adversos , Bloqueadores dos Canais de Cálcio/uso terapêutico , Sinalização do Cálcio/genética , Caveolina 1/genética , Proliferação de Células/efeitos dos fármacos , Hipertensão Pulmonar Primária Familiar/tratamento farmacológico , Hipertensão Pulmonar Primária Familiar/patologia , Feminino , Células HEK293 , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Proteínas do Tecido Nervoso/genética , Canais de Potássio de Domínios Poros em Tandem/genética , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Transdução de Sinais/genética , Sequenciamento do Exoma
3.
Mol Cell ; 48(5): 681-91, 2012 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-23103253

RESUMO

The mammalian target of rapamycin (mTOR) pathway, which is essential for cell proliferation, is repressed in certain cell types in hypoxia. However, hypoxia-inducible factor 2α (HIF2α) can act as a proliferation-promoting factor in some biological settings. This paradoxical situation led us to study whether HIF2α has a specific effect on mTORC1 regulation. Here we show that activation of the HIF2α pathway increases mTORC1 activity by upregulating expression of the amino acid carrier SLC7A5. At the molecular level we also show that HIF2α binds to the Slc7a5 proximal promoter. Our findings identify a link between the oxygen-sensing HIF2α pathway and mTORC1 regulation, revealing the molecular basis of the tumor-promoting properties of HIF2α in von Hippel-Lindau-deficient cells. We also describe relevant physiological scenarios, including those that occur in liver and lung tissue, wherein HIF2α or low-oxygen tension drive mTORC1 activity and SLC7A5 expression.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Carcinoma de Células Renais/metabolismo , Neoplasias Renais/metabolismo , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Proteínas/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Sítios de Ligação , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Hipóxia Celular , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Neoplasias Renais/genética , Neoplasias Renais/patologia , Transportador 1 de Aminoácidos Neutros Grandes/genética , Fígado/metabolismo , Pulmão/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Camundongos SCID , Complexos Multiproteicos , Transplante de Neoplasias , Regiões Promotoras Genéticas , Proteínas/genética , Interferência de RNA , Transdução de Sinais , Serina-Treonina Quinases TOR , Fatores de Tempo , Transfecção , Carga Tumoral , Regulação para Cima , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
4.
Physiology (Bethesda) ; 33(5): 328-337, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30109825

RESUMO

Hypoxia signaling in the vasculature controls vascular permeability, inflammation, vascular growth, and repair of vascular injury. In this review, we summarize recent insights in this burgeoning field and highlight the importance of studying the heterogeneity of hypoxia responses among individual patients, distinct vascular beds, and even individual vascular cells.


Assuntos
Células Endoteliais/fisiologia , Homeostase/fisiologia , Hipóxia/fisiopatologia , Transdução de Sinais/fisiologia , Animais , Permeabilidade Capilar/fisiologia , Humanos
5.
Circulation ; 135(25): 2505-2523, 2017 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-28381471

RESUMO

BACKGROUND: The mechanisms underlying the dedifferentiation and lineage conversion of adult human fibroblasts into functional endothelial cells have not yet been fully defined. Furthermore, it is not known whether fibroblast dedifferentiation recapitulates the generation of multipotent progenitors during embryonic development, which give rise to endothelial and hematopoietic cell lineages. Here we established the role of the developmental transcription factor SOX17 in regulating the bilineage conversion of fibroblasts by the generation of intermediate progenitors. METHODS: CD34+ progenitors were generated after the dedifferentiation of human adult dermal fibroblasts by overexpression of pluripotency transcription factors. Sorted CD34+ cells were transdifferentiated into induced endothelial cells and induced erythroblasts using lineage-specific growth factors. The therapeutic potential of the generated cells was assessed in an experimental model of myocardial infarction. RESULTS: Induced endothelial cells expressed specific endothelial cell surface markers and also exhibited the capacity for cell proliferation and neovascularization. Induced erythroblasts expressed erythroid surface markers and formed erythroid colonies. Endothelial lineage conversion was dependent on the upregulation of the developmental transcription factor SOX17, whereas suppression of SOX17 instead directed the cells toward an erythroid fate. Implantation of these human bipotential CD34+ progenitors into nonobese diabetic/severe combined immunodeficiency (NOD-SCID) mice resulted in the formation of microvessels derived from human fibroblasts perfused with mouse and human erythrocytes. Endothelial cells generated from human fibroblasts also showed upregulation of telomerase. Cell implantation markedly improved vascularity and cardiac function after myocardial infarction without any evidence of teratoma formation. CONCLUSIONS: Dedifferentiation of fibroblasts to intermediate CD34+ progenitors gives rise to endothelial cells and erythroblasts in a SOX17-dependent manner. These findings identify the intermediate CD34+ progenitor state as a critical bifurcation point, which can be tuned to generate functional blood vessels or erythrocytes and salvage ischemic tissue.


Assuntos
Antígenos CD34/fisiologia , Desdiferenciação Celular/fisiologia , Células Endoteliais/fisiologia , Eritroblastos/fisiologia , Fibroblastos/fisiologia , Fatores de Transcrição SOXF/fisiologia , Células-Tronco/fisiologia , Animais , Células Cultivadas , Humanos , Recém-Nascido , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
6.
Circ Res ; 112(5): 802-15, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23334860

RESUMO

RATIONALE: Closure of the ductus arteriosus (DA) is essential for the transition from fetal to neonatal patterns of circulation. Initial PO2-dependent vasoconstriction causes functional DA closure within minutes. Within days a fibrogenic, proliferative mechanism causes anatomic closure. Though modulated by endothelial-derived vasodilators and constrictors, O2 sensing is intrinsic to ductal smooth muscle cells and oxygen-induced DA constriction persists in the absence of endothelium, endothelin, and cyclooxygenase mediators. O2 increases mitochondrial-derived H2O2, which constricts ductal smooth muscle cells by raising intracellular calcium and activating rho kinase. However, the mechanism by which oxygen changes mitochondrial function is unknown. OBJECTIVE: The purpose of this study was to determine whether mitochondrial fission is crucial for O2-induced DA constriction and closure. METHODS AND RESULTS: Using DA harvested from 30 term infants during correction of congenital heart disease, as well as DA from term rabbits, we demonstrate that mitochondrial fission is crucial for O2-induced constriction and closure. O2 rapidly (<5 minutes) causes mitochondrial fission by a cyclin-dependent kinase- mediated phosphorylation of dynamin-related protein 1 (Drp1) at serine 616. Fission triggers a metabolic shift in the ductal smooth muscle cells that activates pyruvate dehydrogenase and increases mitochondrial H2O2 production. Subsequently, fission increases complex I activity. Mitochondrial-targeted catalase overexpression eliminates PO2-induced increases in mitochondrial-derived H2O2 and cytosolic calcium. The small molecule Drp1 inhibitor, Mdivi-1, and siDRP1 yield concordant results, inhibiting O2-induced constriction (without altering the response to phenylephrine or KCl) and preventing O2-induced increases in oxidative metabolism, cytosolic calcium, and ductal smooth muscle cells proliferation. Prolonged Drp1 inhibition reduces DA closure in a tissue culture model. CONCLUSIONS: Mitochondrial fission is an obligatory, early step in mammalian O2 sensing and offers a promising target for modulating DA patency.


Assuntos
Canal Arterial/fisiologia , GTP Fosfo-Hidrolases/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Dinâmica Mitocondrial/fisiologia , Proteínas Mitocondriais/fisiologia , Músculo Liso Vascular/fisiologia , Oxigênio/fisiologia , Vasoconstrição/fisiologia , Animais , Animais Recém-Nascidos , Cálcio/metabolismo , Proliferação de Células , Células Cultivadas , Canal Arterial/citologia , Dinaminas , Feminino , Humanos , Peróxido de Hidrogênio/metabolismo , Recém-Nascido , Masculino , Mitocôndrias/metabolismo , Modelos Animais , Músculo Liso Vascular/citologia , Consumo de Oxigênio/fisiologia , Coelhos , Técnicas de Cultura de Tecidos , Quinases Associadas a rho/metabolismo
7.
Circ Res ; 110(11): 1484-97, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22511751

RESUMO

RATIONALE: Pulmonary arterial hypertension (PAH) is a lethal syndrome characterized by pulmonary vascular obstruction caused, in part, by pulmonary artery smooth muscle cell (PASMC) hyperproliferation. Mitochondrial fragmentation and normoxic activation of hypoxia-inducible factor-1α (HIF-1α) have been observed in PAH PASMCs; however, their relationship and relevance to the development of PAH are unknown. Dynamin-related protein-1 (DRP1) is a GTPase that, when activated by kinases that phosphorylate serine 616, causes mitochondrial fission. It is, however, unknown whether mitochondrial fission is a prerequisite for proliferation. OBJECTIVE: We hypothesize that DRP1 activation is responsible for increased mitochondrial fission in PAH PASMCs and that DRP1 inhibition may slow proliferation and have therapeutic potential. METHODS AND RESULTS: Experiments were conducted using human control and PAH lungs (n=5) and PASMCs in culture. Parallel experiments were performed in rat lung sections and PASMCs and in rodent PAH models induced by the HIF-1α activator, cobalt, chronic hypoxia, and monocrotaline. HIF-1α activation in human PAH leads to mitochondrial fission by cyclin B1/CDK1-dependent phosphorylation of DRP1 at serine 616. In normal PASMCs, HIF-1α activation by CoCl(2) or desferrioxamine causes DRP1-mediated fission. HIF-1α inhibition reduces DRP1 activation, prevents fission, and reduces PASMC proliferation. Both the DRP1 inhibitor Mdivi-1 and siDRP1 prevent mitotic fission and arrest PAH PASMCs at the G2/M interphase. Mdivi-1 is antiproliferative in human PAH PASMCs and in rodent models. Mdivi-1 improves exercise capacity, right ventricular function, and hemodynamics in experimental PAH. CONCLUSIONS: DRP-1-mediated mitotic fission is a cell-cycle checkpoint that can be therapeutically targeted in hyperproliferative disorders such as PAH.


Assuntos
Proliferação de Células , Dinaminas/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Hipertensão Pulmonar/enzimologia , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias Musculares/enzimologia , Proteínas Mitocondriais/metabolismo , Mitose , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Animais , Anti-Hipertensivos/farmacologia , Proteína Quinase CDC2/metabolismo , Estudos de Casos e Controles , Pontos de Checagem do Ciclo Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cobalto , Ciclina B1/metabolismo , Modelos Animais de Doenças , Dinaminas/genética , Ativação Enzimática , Hipertensão Pulmonar Primária Familiar , GTP Fosfo-Hidrolases/genética , Terapia Genética/métodos , Glicólise , Humanos , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/terapia , Hipóxia/complicações , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Proteínas Associadas aos Microtúbulos/genética , Mitocôndrias Musculares/efeitos dos fármacos , Mitocôndrias Musculares/patologia , Proteínas Mitocondriais/genética , Mitose/efeitos dos fármacos , Monocrotalina , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Fosforilação , Artéria Pulmonar/enzimologia , Artéria Pulmonar/patologia , Quinazolinonas/farmacologia , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Serina , Fatores de Tempo , Transfecção
8.
Am J Respir Crit Care Med ; 187(8): 865-78, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23449689

RESUMO

RATIONALE: Pulmonary arterial hypertension (PAH) is a lethal, female-predominant, vascular disease. Pathologic changes in PA smooth muscle cells (PASMC) include excessive proliferation, apoptosis-resistance, and mitochondrial fragmentation. Activation of dynamin-related protein increases mitotic fission and promotes this proliferation-apoptosis imbalance. The contribution of decreased fusion and reduced mitofusin-2 (MFN2) expression to PAH is unknown. OBJECTIVES: We hypothesize that decreased MFN2 expression promotes mitochondrial fragmentation, increases proliferation, and impairs apoptosis. The role of MFN2's transcriptional coactivator, peroxisome proliferator-activated receptor γ coactivator 1-α (PGC1α), was assessed. MFN2 therapy was tested in PAH PASMC and in models of PAH. METHODS: Fusion and fission mediators were measured in lungs and PASMC from patients with PAH and female rats with monocrotaline or chronic hypoxia+Sugen-5416 (CH+SU) PAH. The effects of adenoviral mitofusin-2 (Ad-MFN2) overexpression were measured in vitro and in vivo. MEASUREMENTS AND MAIN RESULTS: In normal PASMC, siMFN2 reduced expression of MFN2 and PGC1α; conversely, siPGC1α reduced PGC1α and MFN2 expression. Both interventions caused mitochondrial fragmentation. siMFN2 increased proliferation. In rodent and human PAH PASMC, MFN2 and PGC1α were decreased and mitochondria were fragmented. Ad-MFN2 increased fusion, reduced proliferation, and increased apoptosis in human PAH and CH+SU. In CH+SU, Ad-MFN2 improved walking distance (381 ± 35 vs. 245 ± 39 m; P < 0.05); decreased pulmonary vascular resistance (0.18 ± 0.02 vs. 0.38 ± 0.14 mm Hg/ml/min; P < 0.05); and decreased PA medial thickness (14.5 ± 0.8 vs. 19 ± 1.7%; P < 0.05). Lung vascularity was increased by MFN2. CONCLUSIONS: Decreased expression of MFN2 and PGC1α contribute to mitochondrial fragmentation and a proliferation-apoptosis imbalance in human and experimental PAH. Augmenting MFN2 has therapeutic benefit in human and experimental PAH.


Assuntos
GTP Fosfo-Hidrolases/deficiência , Proteínas de Choque Térmico/deficiência , Hipertensão Pulmonar/fisiopatologia , Dinâmica Mitocondrial/fisiologia , Proteínas Mitocondriais/deficiência , Fatores de Transcrição/deficiência , Animais , Apoptose/fisiologia , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Tolerância ao Exercício/efeitos dos fármacos , Hipertensão Pulmonar Primária Familiar , Feminino , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/patologia , Pulmão/citologia , Pulmão/patologia , Proteínas de Membrana/administração & dosagem , Proteínas de Membrana/deficiência , Dinâmica Mitocondrial/genética , Proteínas Mitocondriais/administração & dosagem , Miócitos de Músculo Liso/patologia , Miócitos de Músculo Liso/fisiologia , Atrofia Óptica Autossômica Dominante/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Ratos , Ratos Sprague-Dawley
9.
J Mol Cell Cardiol ; 64: 124-31, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24090675

RESUMO

Successful implantation and long-term survival of engineered tissue grafts hinges on adequate vascularization of the implant. Endothelial cells are essential for patterning vascular structures, but they require supportive mural cells such as pericytes/mesenchymal stem cells (MSCs) to generate stable, functional blood vessels. While there is evidence that the angiogenic effect of MSCs is mediated via the secretion of paracrine signals, the identity of these signals is unknown. By utilizing two functionally distinct human MSC clones, we found that so-called "pericytic" MSCs secrete the pro-angiogenic vascular guidance molecule SLIT3, which guides vascular development by directing ROBO4-positive endothelial cells to form networks in engineered tissue. In contrast, "non-pericytic" MSCs exhibit reduced activation of the SLIT3/ROBO4 pathway and do not support vascular networks. Using live cell imaging of organizing 3D vascular networks, we show that siRNA knockdown of SLIT3 in MSCs leads to disorganized clustering of ECs. Knockdown of its receptor ROBO4 in ECs abolishes the generation of functional human blood vessels in an in vivo xenogenic implant. These data suggest that the SLIT3/ROBO4 pathway is required for MSC-guided vascularization in engineered tissues. Heterogeneity of SLIT3 expression may underlie the variable clinical success of MSCs for tissue repair applications.


Assuntos
Proteínas de Membrana/genética , Neovascularização Fisiológica/genética , Receptores de Superfície Celular/genética , Engenharia Tecidual , Ativação Transcricional , Animais , Comunicação Celular , Movimento Celular , Análise por Conglomerados , Células Endoteliais/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Humanos , Proteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Pericitos/citologia , Pericitos/metabolismo , Fenótipo , Interferência de RNA , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Alicerces Teciduais
10.
FASEB J ; 26(5): 2175-86, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22321727

RESUMO

Mitochondria exist in dynamic networks that undergo fusion and fission. Mitochondrial fusion and fission are mediated by several GTPases in the outer mitochondrial membrane, notably mitofusin-2 (Mfn-2), which promotes fusion, and dynamin-related protein (Drp-1), which promotes fission. We report that human lung cancer cell lines exhibit an imbalance of Drp-1/Mfn-2 expression, which promotes a state of mitochondrial fission. Lung tumor tissue samples from patients demonstrated a similar increase in Drp-1 and decrease in Mfn-2 when compared to adjacent healthy lung. Complementary approaches to restore mitochondrial network formation in lung cancer cells by overexpression of Mfn-2, Drp-1 inhibition, or Drp-1 knockdown resulted in a marked reduction of cancer cell proliferation and an increase in spontaneous apoptosis. The number of cancer cells in S phase decreased from 32.4 ± 0.6 to 6.4 ± 0.3% with Drp-1 inhibition (P<0.001). In a xenotransplantation model, Mfn-2 gene therapy or Drp-1 inhibition could regress tumor growth. The tumor volume decreased from 205.6 ± 59 to 70.6 ± 15 mm(3) (P<0.05) with Mfn-2 overexpression and from 186.0 ± 19 to 87.0 ± 6 mm(3) (P<0.01) with therapeutic Drp-1 inhibition. Impaired fusion and enhanced fission contribute fundamentally to the proliferation/apoptosis imbalance in cancer and constitute promising novel therapeutic targets.


Assuntos
Ciclo Celular , Neoplasias Pulmonares/patologia , Mitocôndrias/fisiologia , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Camundongos , Camundongos Nus , Tomografia por Emissão de Pósitrons , Reação em Cadeia da Polimerase em Tempo Real , Tomografia Computadorizada por Raios X
11.
Am J Respir Crit Care Med ; 185(6): 670-9, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22246173

RESUMO

RATIONALE: Pulmonary arterial hypertension (PAH) is a proliferative arteriopathy associated with glucose transporter-1 (Glut1) up-regulation and a glycolytic shift in lung metabolism. Glycolytic metabolism can be detected with the positron emission tomography (PET) tracer (18)F-fluorodeoxyglucose (FDG). OBJECTIVES: The precise cell type in which glycolytic abnormalities occur in PAH is unknown. Moreover, whether FDG-PET is sufficiently sensitive to monitor PAH progression and detect therapeutic regression is untested. We hypothesized that increased lung FDG-PET reflects enhanced glycolysis in vascular cells and is reversible in response to effective therapies. METHODS: PAH was induced in Sprague-Dawley rats by monocrotaline or chronic hypoxia (10% oxygen) in combination with Sugen 5416. Monocrotaline rats were treated with oral dichloroacetate or daily imatinib injections. FDG-PET scans and pulmonary artery acceleration times were obtained weekly. The origin of the PET signal was assessed by laser capture microdissection of airway versus vascular tissue. Metabolism was measured in pulmonary artery smooth muscle cell (PASMC) cultures, using a Seahorse extracellular flux analyzer. MEASUREMENTS AND MAIN RESULTS: Lung FDG increases 1-2 weeks after monocrotaline (when PAH is mild) and is normalized by dichloroacetate and imatinib, which both also regress medial hypertrophy. Glut1 mRNA is up-regulated in both endothelium and PASMCs, but not airway cells or macrophages. PASMCs from monocrotaline rats are hyperproliferative and display normoxic activation of hypoxia-inducible factor-1α (HIF-1α), which underlies their glycolytic phenotype. CONCLUSIONS: HIF-1α-mediated Glut1 up-regulation in proliferating vascular cells in PAH accounts for increased lung FDG-PET uptake. FDG-PET is sensitive to mild PAH and can monitor therapeutic changes in the vasculature.


Assuntos
Fluordesoxiglucose F18 , Hipertensão Pulmonar/diagnóstico por imagem , Monitorização Fisiológica/métodos , Tomografia por Emissão de Pósitrons/métodos , Pressão Propulsora Pulmonar/fisiologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Hipertensão Pulmonar Primária Familiar , Fluordesoxiglucose F18/farmacocinética , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/fisiopatologia , Consumo de Oxigênio , Compostos Radiofarmacêuticos/farmacocinética , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes
12.
Handb Exp Pharmacol ; 218: 105-49, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24092338

RESUMO

World Health Organization category 1 pulmonary hypertension (PH) is a heterogeneous syndrome in which PH originates in the small pulmonary arteries and is therefore also referred to as pulmonary arterial hypertension (PAH). Common pathophysiologic features include endothelial dysfunction, excessive proliferation and impaired apoptosis of vascular cells, and mitochondrial fragmentation. The proliferation/apoptosis imbalance relates in part to activation of the transcription factors hypoxia-inducible factor-1α (HIF-1α) and nuclear factor of activated T-cells (NFAT) and apoptosis repressors, such as survivin. Perivascular inflammation, disruption of adventitial connective tissue, and a glycolytic metabolic shift in vascular cells and right ventricular myocytes also occur in PAH. There are important genetic and epigenetic predispositions to PAH. This review assesses the fidelity of existing animal models to human PAH. No single model can perfectly recapitulate the many diverse forms of PH in Category 1; however, acceptable models exist. PAH induced by monocrotaline and chronic hypoxia plus SU-5416 (CH+SU) in rats display endothelial dysfunction, proliferation/apoptosis imbalance, and develop the glycolytic metabolic profile of human PAH. Histologically, CH+SU best conforms to PAH in that it develops complex vascular lesions, including plexiform lesions. However, the monocrotaline model can be induced to manifest complex vascular lesions and does manifest the tendency of PAH patients to die of right ventricular (RV) failure. Murine models offer greater molecular certainty than rat models but rarely develop significant PH, have less right ventricular hypertrophy (RVH) and pulmonary artery (PA) remodeling, and are harder to image and catheterize. The use of high fidelity catheterization and advanced imaging (microPET-CT, high frequency echocardiography, high field strength MRI) and functional testing (treadmill) permit accurate phenotyping of experimental models of PAH. Preclinical trial design is an important aspect of testing experimental PAH therapies. The use of multiple complementary models with adequate sample size and trial duration and appropriate endpoints are required for preclinical assessment of experimental PAH therapies.


Assuntos
Modelos Animais de Doenças , Hipertensão Pulmonar/etiologia , Animais , Ecocardiografia , Hemodinâmica , Humanos , Hipertensão Pulmonar/fisiopatologia , Hipertensão Pulmonar/terapia , Camundongos , Ratos
13.
Am J Respir Crit Care Med ; 183(8): 1080-91, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21148721

RESUMO

RATIONALE: The etiology of hepatopulmonary syndrome (HPS), a common complication of cirrhosis, is unknown. Inflammation and macrophage accumulation occur in HPS; however, their importance is unclear. Common bile duct ligation (CBDL) creates an accepted model of HPS, allowing us to investigate the cause of HPS. OBJECTIVES: We hypothesized that macrophages are central to HPS and investigated the therapeutic potential of macrophage depletion. METHODS: Hemodynamics, alveolar-arterial gradient, vascular reactivity, and histology were assessed in CBDL versus sham rats (n = 21 per group). The effects of plasma on smooth muscle cell proliferation and endothelial tube formation were measured. Macrophage depletion was used to prevent (gadolinium) or regress (clodronate) HPS. CD68(+) macrophages and capillary density were measured in the lungs of patients with cirrhosis versus control patients (n = 10 per group). MEASUREMENTS AND MAIN RESULTS: CBDL increased cardiac output and alveolar-arterial gradient by causing capillary dilatation and arteriovenous malformations. Activated CD68(+)macrophages (nuclear factor-κB+) accumulated in HPS pulmonary arteries, drawn by elevated levels of plasma endotoxin and lung monocyte chemoattractant protein-1. These macrophages expressed inducible nitric oxide synthase, vascular endothelial growth factor, and platelet-derived growth factor. HPS plasma increased endothelial tube formation and pulmonary artery smooth muscle cell proliferation. Macrophage depletion prevented and reversed the histological and hemodynamic features of HPS. CBDL lungs demonstrated increased medial thickness and obstruction of small pulmonary arteries. Nitric oxide synthase inhibition unmasked exaggerated pulmonary vasoconstrictor responses in HPS. Patients with cirrhosis had increased pulmonary intravascular macrophage accumulation and capillary density. CONCLUSIONS: HPS results from intravascular accumulation of CD68(+)macrophages. An occult proliferative vasculopathy may explain the occasional transition to portopulmonary hypertension. Macrophage depletion may have therapeutic potential in HPS.


Assuntos
Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Síndrome Hepatopulmonar/imunologia , Macrófagos/imunologia , Animais , Antígenos CD/fisiologia , Antígenos de Diferenciação Mielomonocítica/fisiologia , Malformações Arteriovenosas/etiologia , Malformações Arteriovenosas/fisiopatologia , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Síndrome Hepatopulmonar/etiologia , Humanos , Pulmão/irrigação sanguínea , Pulmão/citologia , Pulmão/imunologia , Macrófagos/fisiologia , Masculino , Músculo Liso Vascular/fisiopatologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/fisiologia , Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Fator de Crescimento Derivado de Plaquetas/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/fisiologia
14.
Circulation ; 121(24): 2661-71, 2010 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-20529999

RESUMO

BACKGROUND: Excessive proliferation and impaired apoptosis of pulmonary artery (PA) smooth muscle cells (PASMCs) contribute to vascular obstruction in patients and fawn-hooded rats (FHRs) with PA hypertension (PAH). Expression and activity of mitochondrial superoxide dismutase-2 (SOD2), the major generator of H(2)O(2), is known to be reduced in PAH; however, the mechanism and therapeutic relevance of this are unknown. METHODS AND RESULTS: SOD2 expression in PASMCs is decreased in PAH patients and FHRs with PAH. FHR PASMCs have higher proliferation and lower apoptosis rates than Sprague-Dawley rat PASMCs. Moreover, FHR PASMCs have hyperpolarized mitochondria, low H(2)O(2) production, and reduced cytoplasmic and mitochondrial redox state. Administration of SOD2 small interfering RNA to normal PASMCs recapitulates the FHR PAH phenotype, hyperpolarizing mitochondria, decreasing H(2)O(2), and inhibiting caspase activity. Conversely, SOD2 overexpression in FHR PASMCs or therapy with the SOD-mimetic metalloporphyrin Mn(III)tetrakis (4-benzoic acid) porphyrin (MnTBAP) reverses the hyperproliferative PAH phenotype. Importantly, SOD-mimetic therapy regresses PAH in vivo. Investigation of the SOD2 gene revealed no mutation, suggesting a possible epigenetic dysregulation. Genomic bisulfite sequencing demonstrates selective hypermethylation of a CpG island in an enhancer region of intron 2 and another in the promoter. Differential methylation occurs selectively in PAs versus aortic SMCs and is reversed by the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine, restoring both SOD2 expression and the ratio of proliferation to apoptosis. Expression of the enzymes that mediate gene methylation, DNA methyltransferases 1 and 3B, is upregulated in FHR lungs. CONCLUSIONS: Tissue-specific, epigenetic SOD2 deficiency initiates and sustains a heritable form of PAH by impairing redox signaling and creating a proliferative, apoptosis-resistant PASMC. SOD augmentation regresses experimental PAH. The discovery of an epigenetic component to PAH may offer new therapeutic targets.


Assuntos
Proliferação de Células , Epigênese Genética/genética , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Mitocôndrias Musculares/enzimologia , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Adenoviridae/genética , Animais , Apoptose , Biomimética , Modelos Animais de Doenças , Epigênese Genética/fisiologia , Humanos , Peróxido de Hidrogênio/metabolismo , Hipertensão Pulmonar/tratamento farmacológico , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Fenótipo , Ratos , Ratos Mutantes , Ratos Sprague-Dawley
15.
Circulation ; 119(3): 408-16, 2009 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-19139381

RESUMO

BACKGROUND: Ventricular expression of phosphodiesterase-5 (PDE5), an enzyme responsible for cGMP catabolism, is increased in human right ventricular hypertrophy, but its role in left ventricular (LV) failure remains incompletely understood. We therefore measured LV PDE5 expression in patients with advanced systolic heart failure and characterized LV remodeling after myocardial infarction in transgenic mice with cardiomyocyte-specific overexpression of PDE5 (PDE5-TG). METHODS AND RESULTS: Immunoblot and immunohistochemistry techniques revealed that PDE5 expression was greater in explanted LVs from patients with dilated and ischemic cardiomyopathy than in control hearts. To evaluate the impact of increased ventricular PDE5 levels on cardiac function, PDE5-TG mice were generated. Confocal and immunoelectron microscopy revealed increased PDE5 expression in cardiomyocytes, predominantly localized to Z-bands. At baseline, myocardial cGMP levels, cell shortening, and calcium handling in isolated cardiomyocytes and LV hemodynamic measurements were similar in PDE5-TG and wild-type littermates. Ten days after myocardial infarction, LV cGMP levels had increased to a greater extent in wild-type mice than in PDE5-TG mice (P<0.05). Ten weeks after myocardial infarction, LV end-systolic and end-diastolic volumes were larger in PDE5-TG than in wild-type mice (57+/-5 versus 39+/-4 and 65+/-6 versus 48+/-4 muL, respectively; P<0.01 for both). LV systolic dysfunction and diastolic dysfunction were more marked in PDE5-TG than in wild-type mice, associated with enhanced hypertrophy and reduced contractile function in isolated cardiomyocytes from remote myocardium. CONCLUSIONS: Increased PDE5 expression predisposes mice to adverse LV remodeling after myocardial infarction. Increased myocardial PDE5 expression in patients with advanced cardiomyopathy may contribute to the development of heart failure and represents an important therapeutic target.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/biossíntese , Regulação Enzimológica da Expressão Gênica/fisiologia , Insuficiência Cardíaca/enzimologia , Infarto do Miocárdio/enzimologia , Remodelação Ventricular/genética , Animais , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Insuficiência Cardíaca/fisiopatologia , Ventrículos do Coração/enzimologia , Ventrículos do Coração/fisiopatologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infarto do Miocárdio/fisiopatologia , Miocárdio/enzimologia , Miocárdio/patologia
16.
Basic Res Cardiol ; 105(2): 169-79, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20020305

RESUMO

Nitric oxide modulates the severity of myocardial ischemia-reperfusion (I/R) injury. We investigated whether cardioselective nitric oxide synthase 3 (NOS3) gene transfer could confer myocardial protection against I/R injury in pigs and examined potential molecular mechanisms. I/R injury was induced by balloon occlusion of the left anterior descending artery for 45 min followed by 4 or 72 h reperfusion. Hemodynamic and pathological changes were measured in pigs in the absence (n = 11) or presence of prior intracoronary retroinfusion of human NOS3 (AdNOS3, 5 x 10(10) PFU, n = 13) or control vector (AdRR5, 5 x 10(10) PFU, n = 11). Retrograde NOS3 gene transfer selectively increased NOS3 expression and NO bioavailability in the area at risk (AAR) without changing endogenous NOS isoform expression. At 4 h R, LV systolic (dP/dt(max)) and diastolic (dP/dt(min)) function was better preserved in AdNOS3- than in AdRR5-injected pigs (2,539 +/- 165 vs. 1,829 +/- 156 mmHg/s, and -2,781 +/- 340 vs. -2,062 +/- 292 mmHg/s, respectively, P < 0.05 for both). Myocardial infarct size (% AAR) was significantly smaller in AdNOS3 than in control and AdRR5 and associated with a significantly greater reduction in cardiac myeloperoxidase activity, a marker of neutrophil infiltration. The latter effects were sustained at 72 h R in a subset of pigs (n = 7). In the AAR, intercellular endothelial adhesion molecule-1 expression and cardiomyocyte apoptosis were significantly lower in AdNOS3. In conclusion, single myocardial NOS3 retroinfusion attenuates I/R injury, and causes a sustained reduction in myocardial infarct size and inflammatory cell infiltration. Gene-based strategies to increase NO bioavailability may have therapeutic potential in myocardial I/R.


Assuntos
Terapia Genética , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Óxido Nítrico Sintase Tipo III/genética , Animais , Apoptose , Células Endoteliais/fisiologia , Hemodinâmica , Leucócitos/fisiologia , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/complicações , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Óxido Nítrico Sintase Tipo III/metabolismo , Distribuição Aleatória , Suínos , Transgenes
17.
Stem Cells ; 26(4): 1017-26, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18258720

RESUMO

Circulating endothelial progenitor cells (EPCs) contribute to neovascularization of ischemic tissues and repair of injured endothelium. The role of bone marrow-derived progenitor cells in hypoxia-induced pulmonary vascular remodeling and their tissue-engineering potential in pulmonary hypertension (PH) remain largely unknown. We studied endogenous mobilization and homing of EPCs in green fluorescent protein bone marrow chimeric mice exposed to chronic hypoxia, a common hallmark of PH. Despite increased peripheral mobilization, as shown by flow cytometry and EPC culture, bone marrow-derived endothelial cell recruitment in remodeling lung vessels was limited. Moreover, transfer of vascular endothelial growth factor receptor-2+/Sca-1+/CXCR-4+-cultured early-outgrowth EPCs failed to reverse PH, suggesting hypoxia-induced functional impairment of transferred EPCs. Chronic hypoxia decreased migration to stromal cell-derived factor-1alpha, adhesion to fibronectin, incorporation into a vascular network, and nitric oxide production (-41%, -29%, -30%, and -32%, respectively, vs. normoxic EPCs; p < .05 for all). The dysfunctional phenotype of hypoxic EPCs significantly impaired their neovascularization capacity in chronic hind limb ischemia, contrary to normoxic EPCs cultured in identical conditions. Mechanisms contributing to EPC dysfunction include reduced integrin alphav and beta1 expression, decreased mitochondrial membrane potential, and enhanced senescence. Novel insights from chronic hypoxia-induced EPC dysfunction may provide important cues for improved future cell repair strategies.


Assuntos
Células Endoteliais/fisiologia , Hipertensão Pulmonar/fisiopatologia , Hipóxia/fisiopatologia , Células-Tronco/patologia , Animais , Células Cultivadas , Doença Crônica , Células Endoteliais/patologia , Células Endoteliais/transplante , Membro Posterior/irrigação sanguínea , Membro Posterior/cirurgia , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/cirurgia , Hipóxia/patologia , Hipóxia/cirurgia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco/fisiologia , Fatores de Tempo
18.
Nat Commun ; 10(1): 2126, 2019 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-31073164

RESUMO

Repair of the endothelial cell barrier after inflammatory injury is essential for tissue fluid homeostasis and normalizing leukocyte transmigration. However, the mechanisms of endothelial regeneration remain poorly understood. Here we show that the endothelial and hematopoietic developmental transcription factor Sox17 promotes endothelial regeneration in the endotoxemia model of endothelial injury. Genetic lineage tracing studies demonstrate that the native endothelium itself serves as the primary source of endothelial cells repopulating the vessel wall following injury. We identify Sox17 as a key regulator of endothelial cell regeneration using endothelial-specific deletion and overexpression of Sox17. Endotoxemia upregulates Hypoxia inducible factor 1α, which in turn transcriptionally activates Sox17 expression. We observe that Sox17 increases endothelial cell proliferation via upregulation of Cyclin E1. Furthermore, endothelial-specific upregulation of Sox17 in vivo enhances lung endothelial regeneration. We conclude that endotoxemia adaptively activates Sox17 expression to mediate Cyclin E1-dependent endothelial cell regeneration and restore vascular homeostasis.


Assuntos
Ciclina E/genética , Endotélio Vascular/fisiopatologia , Endotoxemia/patologia , Proteínas HMGB/metabolismo , Proteínas Oncogênicas/genética , Regeneração/imunologia , Fatores de Transcrição SOXF/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Ciclina E/metabolismo , Modelos Animais de Doenças , Células Endoteliais/fisiologia , Endotoxemia/imunologia , Células HEK293 , Proteínas HMGB/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Oncogênicas/metabolismo , Regiões Promotoras Genéticas/genética , Fatores de Transcrição SOXF/genética , Transdução de Sinais/fisiologia , Regulação para Cima
19.
Circulation ; 116(8): 936-43, 2007 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-17679618

RESUMO

BACKGROUND: Nitric oxide (NO) activates soluble guanylate cyclase (sGC), a heterodimer composed of alpha- and beta-subunits, to produce cGMP. NO reduces pulmonary vascular remodeling, but the role of sGC in vascular responses to acute and chronic hypoxia remains incompletely elucidated. We therefore studied pulmonary vascular responses to acute and chronic hypoxia in wild-type (WT) mice and mice with a nonfunctional alpha1-subunit (sGCalpha1-/-). METHODS AND RESULTS: sGCalpha1-/- mice had significantly reduced lung sGC activity and vasodilator-stimulated phosphoprotein phosphorylation. Right ventricular systolic pressure did not differ between genotypes at baseline and increased similarly in WT (22+/-2 to 34+/-2 mm Hg) and sGCalpha1-/- (23+/-2 to 34+/-1 mm Hg) mice in response to acute hypoxia. Inhaled NO (40 ppm) blunted the increase in right ventricular systolic pressure in WT mice (22+/-2 to 24+/-2 mm Hg, P<0.01 versus hypoxia without NO) but not in sGCalpha1-/- mice (22+/-1 to 33+/-1 mm Hg) and was accompanied by a significant rise in lung cGMP content only in WT mice. In contrast, the NO-donor sodium nitroprusside (1.5 mg/kg) decreased systemic blood pressure similarly in awake WT and sGCalpha1-/- mice as measured by telemetry (-37+/-2 versus -42+/-4 mm Hg). After 3 weeks of hypoxia, the increases in right ventricular systolic pressure, right ventricular hypertrophy, and muscularization of intra-acinar pulmonary vessels were 43%, 135%, and 46% greater, respectively, in sGCalpha1-/- than in WT mice (P<0.01). Increased remodeling in sGCalpha1-/- mice was associated with an increased frequency of 5'-bromo-deoxyuridine-positive vessels after 1 and 3 weeks (P<0.01 versus WT). CONCLUSIONS: Deficiency of sGCalpha1 does not alter hypoxic pulmonary vasoconstriction. sGCalpha1 is essential for NO-mediated pulmonary vasodilation and limits chronic hypoxia-induced pulmonary vascular remodeling.


Assuntos
Guanilato Ciclase/genética , Guanilato Ciclase/metabolismo , Hipertensão Pulmonar/fisiopatologia , Hipóxia/fisiopatologia , Óxido Nítrico/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Vasodilatação/fisiologia , Doença Aguda , Animais , Antimetabólitos/farmacocinética , Pressão Sanguínea/fisiologia , Bromodesoxiuridina/farmacocinética , Doença Crônica , GMP Cíclico/metabolismo , Dimerização , Feminino , Guanilato Ciclase/química , Hipertensão Pulmonar/metabolismo , Hipertrofia Ventricular Direita/metabolismo , Hipertrofia Ventricular Direita/fisiopatologia , Hipóxia/metabolismo , Masculino , Camundongos , Camundongos Mutantes , Artéria Pulmonar/fisiologia , Circulação Pulmonar/fisiologia , Receptores Citoplasmáticos e Nucleares/química , Guanilil Ciclase Solúvel , Função Ventricular Direita/fisiologia
20.
Mol Biol Cell ; 2017 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-28298490

RESUMO

A heterozygous Caveolin-1 c.474delA mutation has been identified in a family with heritable pulmonary arterial hypertension (PAH). This frameshift mutation leads to caveolin-1 protein that contains all known functional domains but has a change only in the final 20 amino acids of the C terminus. Here we studied how this mutation alters caveolin-1 function using patient-derived fibroblasts. Transmission electron microscopy showed that fibroblasts carrying the c.474delA mutation formed typical caveolae. Expression of mutated caveolin-1 in caveolin-1-null mouse fibroblasts failed to induce formation of caveolae due to retention of the mutated protein in the endoplasmic reticulum. However, co-expression of wild type caveolin-1 with mutated caveolin-1 restored the ability to form caveolae. Importantly, fibroblasts carrying the mutation showed 2-fold increase in proliferation rate associated with hyper-phosphorylation of Smad1/5/8. This mutation impaired the anti-proliferative function of caveolin-1. Inhibition of type I TGFß receptors ALK1/2/3/6 responsible for phosphorylation of Smad1/5/8 reduced the hyper-proliferation seen in c.474delA fibroblasts. These results demonstrate the critical role of the final 20 amino acids of caveolin-1 in modulating fibroblast proliferation through dampening Smad signaling, and suggest that augmented Smad signaling and fibroblast hyper-proliferation are contributing factors in the pathogenesis of PAH in patients with caveolin-1 c.474delA mutation.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa