Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
BMC Neurol ; 22(1): 469, 2022 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-36494773

RESUMO

BACKGROUND: Hereditary transthyretin (ATTRv) amyloidosis is a rare, genetically heterogeneous and phenotypically variable systemic disease characterized by deposition of misfolded transthyretin fibrils in various tissues. ATTRv cardiomyopathy and progressive axonal polyneuropathy are the most common manifestations, leading to severe disability and ultimately death within approximately ten years. As disease-modifying treatment options evolve, timely diagnosis and treatment initiation are crucial to prevent rapid disease progression. CASE PRESENTATION: Here, we report on a 73-year old patient initially diagnosed with cardiac wild-type ATTR (ATTRwt) amyloidosis by endomyocardial biopsy. Molecular genetic analysis revealed a novel TTR sequence variant (p.Ala65Val) that is highly likely to be amyloidogenic in light of previously reported TTR mutations and the patient's clinical presentation and family history. CONCLUSIONS: Our findings expand the spectrum of known pathogenic TTR mutations and underline the importance of a thorough diagnostic workup in amyloidosis patients including careful genetic testing to avoid misdiagnosis and missing of treatment opportunities and to enable cascade testing and tracking of carriers.


Assuntos
Neuropatias Amiloides Familiares , Cardiomiopatias , Humanos , Neuropatias Amiloides Familiares/complicações , Neuropatias Amiloides Familiares/diagnóstico , Neuropatias Amiloides Familiares/genética , Mutação/genética , Cardiomiopatias/diagnóstico , Cardiomiopatias/genética , Fenótipo , Progressão da Doença
2.
Parkinsonism Relat Disord ; 90: 73-78, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34399161

RESUMO

INTRODUCTION: Although there has been increasing recognition of the occurrence of non-epileptic involuntary movements in developmental and epileptic encephalopathies (DEEs), the spectrum of dystonic presentations associated with these conditions remains poorly described. We sought to expand the catalogue of dystonia-predominant phenotypes in monogenic DEEs, building on the recently introduced concept of an epilepsy-movement disorder spectrum. METHODS: Cases were identified from a whole-exome-sequenced cohort of 45 pediatric index patients with complex dystonia (67% sequenced as parent-child trios). Review of molecular findings in DEE-associated genes was performed. For five individuals with identified DEE-causing variants, detailed information about presenting phenotypic features and the natural history of disease was obtained. RESULTS: De-novo pathogenic and likely pathogenic missense variants in GABRA1, GABBR2, GNAO1, and FOXG1 gave rise to infantile-onset persistent and paroxysmal dystonic manifestations, beginning in the limb or truncal musculature and progressing gradually to a generalized state. Coexisting, less prominent movement-disorder symptoms were observed and included myoclonic, ballistic, and stereotypic abnormal movements as well as choreoathetosis. Dystonia dominated over epileptic neurodevelopmental comorbidities in all four subjects and represented the primary indication for molecular genetic analysis. We also report the unusual case of an adult female patient with dystonia, tremor, and mild learning disability who was found to harbor a pathogenic frameshift variant in MECP2. CONCLUSIONS: Dystonia can be a leading clinical manifestation in different DEEs. A monogenic basis of disease should be considered on the association of dystonia and developmental delay-epilepsy presentations, justifying a molecular screening for variants in DEE-associated genes.


Assuntos
Encefalopatias/genética , Distonia/genética , Síndromes Epilépticas/genética , Transtornos do Neurodesenvolvimento/genética , Adolescente , Encefalopatias/complicações , Criança , Pré-Escolar , Síndromes Epilépticas/complicações , Feminino , Fatores de Transcrição Forkhead/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Humanos , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Proteínas do Tecido Nervoso/genética , Transtornos do Neurodesenvolvimento/complicações , Fenótipo , Receptores de GABA-A/genética , Receptores de GABA-B/genética
3.
Cardiovasc Diagn Ther ; 11(2): 637-649, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33968641

RESUMO

BACKGROUND: Comprehensive genetic analysis yields in a higher diagnostic rate but also in a higher number of secondary findings (SF). American College of Medical Genetics and Genomics (ACMG) published a list of 59 actionable genes for which disease causing sequence variants are recommended to be reported as SF including 27 genes linked to inherited cardiovascular disease (CVD) such as arrhythmia syndromes, cardiomyopathies and vascular and connective tissue disorders. One of the selected conditions represented in the actionable gene list is the arrhythmogenic right ventricle cardiomyopathy (ARVC), an inherited heart muscle disease with a particularly high risk of sudden cardiac death (SCD). Since clinical symptoms are frequently absent before SCD, a genetic finding is a promising option for early diagnosis and possible intervention. However, the variant interpretation and the decision to return a SF is still challenging. METHODS: To determine the frequency of medically actionable SF linked to CVD we analyzed data of 6,605 individuals who underwent high throughput sequencing for noncardiac diagnostic requests. In particular, we critically assessed and classified the variants in the ARVC genes: DSC2, DSG2, DSP, PKP2 and TMEM43 and compared our findings with the population-based genome Aggregation Database (gnomAD) and ARVC-afflicted individuals listed in ClinVar and ARVC database. RESULTS: 1% (69/6,605) of tested individuals carried pathogenic SF in one of the 27 genes linked to CVD, of them 13 individuals (0.2%) carried a pathogenic SF in a ARVC gene. Overall, 582 rare variants were identified in all five ARVC genes, 96% of the variants were missense variants and 4% putative LoF variants (pLoF): frameshift, start/stop-gain/loss, splice-site. Finally, we selected 13 of the 24 pLoF variants as pathogenic SF by careful data interpretation. CONCLUSIONS: Since SF in actionable ARVC genes can allow early detection and prevention of disease and SCD, detected variant must undergo rigorous clinical and laboratory evaluation before it can be described as pathogenic and returned to patients. Returning a SF to a patient should be interdisciplinary, it needs genetic counselling and clinicians experienced in inherited heart disease.

4.
BMC Med Genomics ; 14(1): 94, 2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33789662

RESUMO

BACKGROUND: Molecular autopsy represents an efficient tool to save the diagnosis in up to one-third of sudden unexplained death (SUD). A defined gene panel is usually used for the examination. Alternatively, it is possible to carry out a comprehensive genetic assessment (whole exome sequencing, WES), which also identifies rare, previously unknown variants. The disadvantage is that a dramatic number of variants must be assessed to identify the causal variant. To improve the evaluation of WES, the human phenotype ontology (HPO) annotation is used internationally for deep phenotyping in the field of rare disease. However, a HPO-based evaluation of WES in SUD has not been described before. METHODS: We performed WES in tissue samples from 16 people after SUD. Instead of a fixed gene panel, we defined a set of HPO terms and thus created a flexible "virtual gene panel", with the advantage, that recently identified genes are automatically associated by HPO terms in the HPO database. RESULTS: We obtained a mean value of 68,947 variants per sample. Stringent filtering ended up in a mean value of 276 variants per sample. Using the HPO-driven virtual gene panel we developed an algorithm that prioritized 1.4% of the variants. Variant interpretation resulted in eleven potentially causative variants in 16 individuals. CONCLUSION: Our data introduce an effective diagnostic procedure in molecular autopsy of SUD with a non-specific clinical phenotype.


Assuntos
Sequenciamento do Exoma , Autopsia , Biologia Computacional , Exoma , Humanos , Masculino , Fenótipo
5.
Neuromuscul Disord ; 31(2): 123-133, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33414056

RESUMO

More than 80 genes are known to be associated with Charcot-Marie-Tooth disease (CMT). Mutations of LRSAM1 were identified as a rare cause and define the subgroup of axonal neuropathy CMT2P. We identified additional 14 patients out of 12 families. Clinical and electrophysiological data confirm a late-onset axonal neuropathy with a predominance of sensorimotor impairment. The patients harbored ten different variants in LRSAM1, seven of which were novel. Due to variable inheritance patterns and clustering of pathogenic variants in 3´-prime exons, interpretation of genetic variants in LRSAM1 is challenging. The majority follows dominant inheritance, whereas recessive inheritance has been described for one variant. Variants at the 3`end may or may not escape from nonsense-mediated decay, thereby defining the pattern of inheritance. Our data emphasize the importance of the C-terminal RING domain, which exerts a dominant-negative effect on protein function, whenever affected by an altered or truncated protein. In conclusion, CMT2P is a rare, but nevertheless relevant cause of adult-onset axonal and painful neuropathy. ACMG (American College of Medical Genetics and genomics) criteria should be carefully applied in variant interpretation, with special attention to premature termination codon-introducing variants and their location within the gene.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Estudos de Associação Genética , Mutação/genética , Ubiquitina-Proteína Ligases , Adolescente , Adulto , Idoso , Axônios/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo , Ubiquitina-Proteína Ligases/genética , Adulto Jovem
6.
Circ Res ; 128(3): 401-418, 2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33322916

RESUMO

RATIONALE: In pulmonary arterial hypertension (PAH), endothelial dysfunction and obliterative vascular disease are associated with DNA damage and impaired signaling of BMPR2 (bone morphogenetic protein type 2 receptor) via two downstream transcription factors, PPARγ (peroxisome proliferator-activated receptor gamma), and p53. OBJECTIVE: We investigated the vasculoprotective and regenerative potential of a newly identified PPARγ-p53 transcription factor complex in the pulmonary endothelium. METHODS AND RESULTS: In this study, we identified a pharmacologically inducible vasculoprotective mechanism in pulmonary arterial and lung MV (microvascular) endothelial cells in response to DNA damage and oxidant stress regulated in part by a BMPR2 dependent transcription factor complex between PPARγ and p53. Chromatin immunoprecipitation sequencing and RNA-sequencing established an inducible PPARγ-p53 mediated regenerative program regulating 19 genes involved in lung endothelial cell survival, angiogenesis and DNA repair including, EPHA2 (ephrin type-A receptor 2), FHL2 (four and a half LIM domains protein 2), JAG1 (jagged 1), SULF2 (extracellular sulfatase Sulf-2), and TIGAR (TP53-inducible glycolysis and apoptosis regulator). Expression of these genes was partially impaired when the PPARγ-p53 complex was pharmacologically disrupted or when BMPR2 was reduced in pulmonary artery endothelial cells (PAECs) subjected to oxidative stress. In endothelial cell-specific Bmpr2-knockout mice unable to stabilize p53 in endothelial cells under oxidative stress, Nutlin-3 rescued endothelial p53 and PPARγ-p53 complex formation and induced target genes, such as APLN (apelin) and JAG1, to regenerate pulmonary microvessels and reverse pulmonary hypertension. In PAECs from BMPR2 mutant PAH patients, pharmacological induction of p53 and PPARγ-p53 genes repaired damaged DNA utilizing genes from the nucleotide excision repair pathway without provoking PAEC apoptosis. CONCLUSIONS: We identified a novel therapeutic strategy that activates a vasculoprotective gene regulation program in PAECs downstream of dysfunctional BMPR2 to rehabilitate PAH PAECs, regenerate pulmonary microvessels, and reverse disease. Our studies pave the way for p53-based vasculoregenerative therapies for PAH by extending the therapeutic focus to PAEC dysfunction and to DNA damage associated with PAH progression.


Assuntos
Indutores da Angiogênese/farmacologia , Células Endoteliais/efeitos dos fármacos , Imidazóis/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , PPAR gama/metabolismo , Piperazinas/farmacologia , Hipertensão Arterial Pulmonar/tratamento farmacológico , Artéria Pulmonar/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Knockout , Estresse Oxidativo , PPAR gama/genética , Hipertensão Arterial Pulmonar/genética , Hipertensão Arterial Pulmonar/metabolismo , Hipertensão Arterial Pulmonar/fisiopatologia , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Artéria Pulmonar/fisiopatologia , Transdução de Sinais , Proteína Supressora de Tumor p53/genética
7.
Hum Mutat ; 41(5): 1025-1032, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32048431

RESUMO

As comprehensive sequencing technologies gain widespread use, questions about so-called secondary findings (SF) require urgent consideration. The American College of Medical Genetics and Genomics has recommended to report SF in 59 genes (ACMG SF v2.0) including four actionable genes associated with inherited primary arrhythmia syndromes (IPAS) such as catecholaminergic polymorphic ventricular tachycardia, long QT syndrome, and Brugada syndrome. Databases provide conflicting results for the purpose of identifying pathogenic variants in SF associated with IPAS at a level of sufficient evidence for clinical return. As IPAS account for a significant proportion of sudden cardiac deaths (SCD) in young and apparently healthy individuals, variant interpretation has a great impact on diagnosis and prevention of disease. Of 6381 individuals, 0.4% carry pathogenic variants in one of the four actionable genes related to IPAS: RYR2, KCNQ1, KCNH2, and SCN5A. Comparison of the databases ClinVar, Leiden Open-source Variant Database, and Human Gene Mutation Database showed impactful differences (0.2% to 1.3%) in variant interpretation improvable by expert-curation depending on database and classification system used. These data further highlight the need for international consensus regarding the variant interpretation, and subsequently management of SF in particular with regard to treatable arrhythmic disorders with increased risk of SCD.


Assuntos
Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Alelos , Bases de Dados Genéticas , Feminino , Estudos de Associação Genética/métodos , Testes Genéticos , Variação Genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Fenótipo , Síndrome
8.
Cell Rep ; 26(5): 1333-1343.e7, 2019 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-30699358

RESUMO

Using proteomic approaches, we uncovered a DNA damage response (DDR) function for peroxisome proliferator activated receptor γ (PPARγ) through its interaction with the DNA damage sensor MRE11-RAD50-NBS1 (MRN) and the E3 ubiquitin ligase UBR5. We show that PPARγ promotes ATM signaling and is essential for UBR5 activity targeting ATM interactor (ATMIN). PPARγ depletion increases ATMIN protein independent of transcription and suppresses DDR-induced ATM signaling. Blocking ATMIN in this context restores ATM activation and DNA repair. We illustrate the physiological relevance of PPARγ DDR functions by using pulmonary arterial hypertension (PAH) as a model that has impaired PPARγ signaling related to endothelial cell (EC) dysfunction and unresolved DNA damage. In pulmonary arterial ECs (PAECs) from PAH patients, we observed disrupted PPARγ-UBR5 interaction, heightened ATMIN expression, and DNA lesions. Blocking ATMIN in PAH PAEC restores ATM activation. Thus, impaired PPARγ DDR functions may explain the genomic instability and loss of endothelial homeostasis in PAH.


Assuntos
Reparo do DNA , Células Endoteliais/metabolismo , Homeostase , PPAR gama/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Dano ao DNA , Instabilidade Genômica , Células HEK293 , Humanos , Modelos Biológicos , Ligação Proteica , Artéria Pulmonar/patologia , Transdução de Sinais , Ubiquitinação
9.
Mol Cell Probes ; 44: 14-20, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30682426

RESUMO

The heterooctameric mitochondrial trifunctional protein (MTP), composed of four α- and ß-subunits harbours three enzymes that each perform a different function in mitochondrial fatty acid ß-oxidation. Pathogenic variants in the MTP genes (HADHA and HADHB) cause MTP deficiency, a rare autosomal recessive metabolic disorder characterized by phenotypic heterogeneity ranging from severe, early-onset, cardiac disease to milder, later-onset, myopathy and neuropathy. Since metabolic myopathies and neuropathies are a group of rare genetic disorders and their associated muscle symptoms may be subtle, the diagnosis is often delayed. Here we evaluated data of 161 patients with myopathy and 242 patients with neuropathy via next generation sequencing (NGS) and report the diagnostic yield in three patients of this cohort by the detection of disease-causing variants in the HADHA or HADHB gene. The mitigated phenotypes of this treatable disease were missed by the newborn screening, highlighting the importance of phenotype-based NGS analysis in patients with rare and clinically very variable disorders such as MTP deficiency.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala/métodos , Subunidade alfa da Proteína Mitocondrial Trifuncional/genética , Subunidade beta da Proteína Mitocondrial Trifuncional/genética , Mutação/genética , Adolescente , Cardiomiopatias/genética , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Humanos , Lactente , Erros Inatos do Metabolismo Lipídico/genética , Masculino , Miopatias Mitocondriais/genética , Proteína Mitocondrial Trifuncional/deficiência , Proteína Mitocondrial Trifuncional/genética , Doenças do Sistema Nervoso/genética , Fenótipo , Rabdomiólise/genética , Síndrome
10.
Mol Genet Genomic Med ; 6(6): 1188-1198, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30406974

RESUMO

BACKGROUND: The diagnosis of mitochondrial disorders is challenging because of the clinical variability and genetic heterogeneity of these conditions. Next-Generation Sequencing (NGS) technology offers a robust high-throughput platform for nuclear and mitochondrial DNA (mtDNA) analyses. METHOD: We developed a custom Agilent SureSelect Mitochondrial and Nuclear Disease Panel (Mito-aND-Panel) capture kit that allows parallel enrichment for subsequent NGS-based sequence analysis of nuclear mitochondrial disease-related genes and the complete mtDNA genome. Sequencing of enriched mtDNA simultaneously with nuclear genes was compared with the separated sequencing of the mitochondrial genome and whole exome sequencing (WES). RESULTS: The Mito-aND-Panel permits accurate detection of low-level mtDNA heteroplasmy due to a very high sequencing depth compared to standard diagnostic procedures using Sanger sequencing/SNaPshot and WES which is crucial to identify maternally inherited mitochondrial disorders. CONCLUSION: We established a NGS-based method with combined sequencing of the complete mtDNA and nuclear genes which enables a more sensitive heteroplasmy detection of mtDNA mutations compared to traditional methods. Because the method promotes the analysis of mtDNA variants in large cohorts, it is cost-effective and simple to setup, we anticipate this is a highly relevant method for sequence-based genetic diagnosis in clinical diagnostic applications.


Assuntos
Testes Genéticos/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Doenças Mitocondriais/genética , Análise de Sequência de DNA/métodos , Custos e Análise de Custo , DNA Mitocondrial/química , DNA Mitocondrial/genética , Testes Genéticos/economia , Testes Genéticos/normas , Sequenciamento de Nucleotídeos em Larga Escala/economia , Sequenciamento de Nucleotídeos em Larga Escala/normas , Humanos , Doenças Mitocondriais/diagnóstico , Sensibilidade e Especificidade , Análise de Sequência de DNA/economia , Análise de Sequência de DNA/normas
11.
Sci Rep ; 8(1): 11682, 2018 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-30076399

RESUMO

Deletions in mitochondrial DNA (mtDNA) are an important cause of human disease and their accumulation has been implicated in the ageing process. As mtDNA is a high copy number genome, the coexistence of deleted and wild-type mtDNA molecules within a single cell defines heteroplasmy. When deleted mtDNA molecules, driven by intracellular clonal expansion, reach a sufficiently high level, a biochemical defect emerges, contributing to the appearance and progression of clinical pathology. Consequently, it is relevant to determine the heteroplasmy levels within individual cells to understand the mechanism of clonal expansion. Heteroplasmy is reflected in a mosaic distribution of cytochrome c oxidase (COX)-deficient muscle fibers. We applied droplet digital PCR (ddPCR) to single muscle fibers collected by laser-capture microdissection (LCM) from muscle biopsies of patients with different paradigms of mitochondrial disease, characterized by the accumulation of single or multiple mtDNA deletions. By combining these two sensitive approaches, ddPCR and LCM, we document different models of clonal expansion in patients with single and multiple mtDNA deletions, implicating different mechanisms and time points for the development of COX deficiency in these molecularly distinct mitochondrial cytopathies.


Assuntos
DNA Mitocondrial/genética , Células Musculares/metabolismo , Reação em Cadeia da Polimerase/métodos , Deleção de Sequência/genética , Adolescente , Adulto , Idoso , Biópsia , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Feminino , GTP Fosfo-Hidrolases/genética , Dosagem de Genes , Genes Recessivos , Humanos , Masculino , Pessoa de Meia-Idade , Fibras Musculares Esqueléticas/metabolismo , Mutação/genética , Fosforilação Oxidativa , Reprodutibilidade dos Testes , Succinato Desidrogenase/metabolismo , Adulto Jovem
13.
Circulation ; 133(18): 1783-94, 2016 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-27045138

RESUMO

BACKGROUND: We previously reported high-throughput RNA sequencing analyses that identified heightened expression of the chromatin architectural factor High Mobility Group AT-hook 1 (HMGA1) in pulmonary arterial endothelial cells (PAECs) from patients who had idiopathic pulmonary arterial hypertension (PAH) in comparison with controls. Because HMGA1 promotes epithelial-to-mesenchymal transition in cancer, we hypothesized that increased HMGA1 could induce transition of PAECs to a smooth muscle (SM)-like mesenchymal phenotype (endothelial-to-mesenchymal transition), explaining both dysregulation of PAEC function and possible cellular contribution to the occlusive remodeling that characterizes advanced idiopathic PAH. METHODS AND RESULTS: We documented increased HMGA1 in PAECs cultured from idiopathic PAH versus donor control lungs. Confocal microscopy of lung explants localized the increase in HMGA1 consistently to pulmonary arterial endothelium, and identified many cells double-positive for HMGA1 and SM22α in occlusive and plexogenic lesions. Because decreased expression and function of bone morphogenetic protein receptor 2 (BMPR2) is observed in PAH, we reduced BMPR2 by small interfering RNA in control PAECs and documented an increase in HMGA1 protein. Consistent with transition of PAECs by HMGA1, we detected reduced platelet endothelial cell adhesion molecule 1 (CD31) and increased endothelial-to-mesenchymal transition markers, αSM actin, SM22α, calponin, phospho-vimentin, and Slug. The transition was associated with spindle SM-like morphology, and the increase in αSM actin was largely reversed by joint knockdown of BMPR2 and HMGA1 or Slug. Pulmonary endothelial cells from mice with endothelial cell-specific loss of Bmpr2 showed similar gene and protein changes. CONCLUSIONS: Increased HMGA1 in PAECs resulting from dysfunctional BMPR2 signaling can transition endothelium to SM-like cells associated with PAH.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/deficiência , Transição Epitelial-Mesenquimal/fisiologia , Proteína HMGA1a/biossíntese , Hipertensão Pulmonar/metabolismo , Fatores de Transcrição da Família Snail/biossíntese , Adolescente , Adulto , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Células Cultivadas , Criança , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Feminino , Proteína HMGA1a/genética , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/patologia , Lactente , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Fatores de Transcrição da Família Snail/genética , Adulto Jovem
14.
Am J Respir Crit Care Med ; 191(11): 1273-86, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25853696

RESUMO

RATIONALE: Pulmonary arterial hypertension is characterized by endothelial dysfunction, impaired bone morphogenetic protein receptor 2 (BMPR2) signaling, and increased elastase activity. Synthetic elastase inhibitors reverse experimental pulmonary hypertension but cause hepatotoxicity in clinical studies. The endogenous elastase inhibitor elafin attenuates hypoxic pulmonary hypertension in mice, but its potential to improve endothelial function and BMPR2 signaling, and to reverse severe experimental pulmonary hypertension or vascular pathology in the human disease was unknown. OBJECTIVES: To assess elafin-mediated regression of pulmonary vascular pathology in rats and in lung explants from patients with pulmonary hypertension. To determine if elafin amplifies BMPR2 signaling in pulmonary artery endothelial cells and to elucidate the underlying mechanism. METHODS: Rats with pulmonary hypertension induced by vascular endothelial growth factor receptor blockade and hypoxia (Sugen/hypoxia) as well as lung organ cultures from patients with pulmonary hypertension were used to assess elafin-mediated reversibility of pulmonary vascular disease. Pulmonary arterial endothelial cells from patients and control subjects were used to determine the efficacy and mechanism of elafin-mediated BMPR2 signaling. MEASUREMENTS AND MAIN RESULTS: In Sugen/hypoxia rats, elafin reduced elastase activity and reversed pulmonary hypertension, judged by regression of right ventricular systolic pressure and hypertrophy and pulmonary artery occlusive changes. Elafin improved endothelial function by increasing apelin, a BMPR2 target. Elafin induced apoptosis in human pulmonary arterial smooth muscle cells and decreased neointimal lesions in lung organ culture. In normal and patient pulmonary artery endothelial cells, elafin promoted angiogenesis by increasing pSMAD-dependent and -independent BMPR2 signaling. This was linked mechanistically to augmented interaction of BMPR2 with caveolin-1 via elafin-mediated stabilization of endothelial surface caveolin-1. CONCLUSIONS: Elafin reverses obliterative changes in pulmonary arteries via elastase inhibition and caveolin-1-dependent amplification of BMPR2 signaling.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/efeitos dos fármacos , Caveolina 1/efeitos dos fármacos , Elafina/farmacologia , Hipertensão Pulmonar/tratamento farmacológico , Inibidores de Proteases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Humanos , Miócitos de Músculo Liso/efeitos dos fármacos , Elastase Pancreática/efeitos dos fármacos , Ratos
15.
Cell Metab ; 21(4): 596-608, 2015 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-25863249

RESUMO

Mitochondrial dysfunction, inflammation, and mutant bone morphogenetic protein receptor 2 (BMPR2) are associated with pulmonary arterial hypertension (PAH), an incurable disease characterized by pulmonary arterial (PA) endothelial cell (EC) apoptosis, decreased microvessels, and occlusive vascular remodeling. We hypothesized that reduced BMPR2 induces PAEC mitochondrial dysfunction, promoting a pro-inflammatory or pro-apoptotic state. Mice with EC deletion of BMPR2 develop hypoxia-induced pulmonary hypertension that, in contrast to non-transgenic littermates, does not reverse upon reoxygenation and is associated with reduced PA microvessels and lung EC p53, PGC1α and TFAM, regulators of mitochondrial biogenesis, and mitochondrial DNA. Decreasing PAEC BMPR2 by siRNA during reoxygenation represses p53, PGC1α, NRF2, TFAM, mitochondrial membrane potential, and ATP and induces mitochondrial DNA deletion and apoptosis. Reducing PAEC BMPR2 in normoxia increases p53, PGC1α, TFAM, mitochondrial membrane potential, ATP production, and glycolysis, and induces mitochondrial fission and a pro-inflammatory state. These features are recapitulated in PAECs from PAH patients with mutant BMPR2.


Assuntos
Sobrevivência Celular/fisiologia , Células Endoteliais/fisiologia , Hipertensão Pulmonar/metabolismo , Mitocôndrias/metabolismo , Modelos Biológicos , Artéria Pulmonar/fisiologia , Regeneração/fisiologia , Análise de Variância , Animais , Western Blotting , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , DNA/metabolismo , Primers do DNA/genética , Citometria de Fluxo , Imunofluorescência , Células HEK293 , Humanos , Hipertensão Pulmonar/fisiopatologia , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Reação em Cadeia da Polimerase , Artéria Pulmonar/citologia , RNA Interferente Pequeno/genética
16.
J Cell Sci ; 125(Pt 4): 956-64, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22399808

RESUMO

Urotensin-II (U-II) has been considered as one of the most potent vasoactive peptides, although its physiological and pathophysiological role is still not finally resolved. Recent evidence suggests that it promotes angiogenic responses in endothelial cells, although the underlying signalling mechanisms are unclear. Reactive oxygen species derived from NADPH oxidases are major signalling molecules in the vasculature. Because NOX2 is functional in endothelial cells, we investigated the role of the NOX2-containing NADPH oxidase in U-II-induced angiogenesis and elucidated a possible contribution of hypoxia-inducible factor-1 (HIF-1), the master regulator of hypoxic angiogenesis, in the response to U-II. We found that U-II increases angiogenesis in vitro and in vivo, and these responses were prevented by antioxidants, NOX2 knockdown and in Nox2(-/-) mice. In addition, U-II-induced angiogenesis was dependent on HIF-1. Interestingly, U-II increased NOX2 transcription involving HIF-1, and chromatin immunoprecipitation confirmed NOX2 as a target gene of HIF-1. In support, NOX2 levels were greatly diminished in U-II-stimulated isolated vessels derived from mice deficient in endothelial HIF-1. Conversely, reactive oxygen species derived from NOX2 were required for U-II activation of HIF and upregulation of HIF-1. In line with this, U-II-induced upregulation of HIF-1 was absent in Nox2(-/-) vessels. Collectively, these findings identified HIF-1 and NOX2 as partners acting in concert to promote angiogenesis in response to U-II. Because U-II has been found to be elevated in cardiovascular disorders and in tumour tissues, this feed-forward mechanism could be an interesting anti-angiogenic therapeutic option in these disorders.


Assuntos
Fator 1 Induzível por Hipóxia/metabolismo , Glicoproteínas de Membrana/metabolismo , NADPH Oxidases/metabolismo , Neovascularização Fisiológica , Urotensinas/metabolismo , Animais , Retroalimentação Fisiológica , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , NADPH Oxidase 2 , NADPH Oxidases/biossíntese , NADPH Oxidases/deficiência , NADPH Oxidases/genética , Neovascularização Patológica , Neovascularização Fisiológica/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Regulação para Cima/efeitos dos fármacos , Urotensinas/farmacologia
17.
Mol Cancer Res ; 10(1): 52-65, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22080479

RESUMO

Ewing tumors comprise the second most common type of bone-associated cancer in children and are characterized by oncogenic EWS/FLI1 fusion proteins and early metastasis. Compelling evidence suggests that elevated levels of intracellular oxidative stress contribute to enhanced aggressiveness of numerous cancers, possibly including Ewing tumors. Using comprehensive microarray analyses and RNA interference, we identified the six-transmembrane epithelial antigen of the prostate 1 (STEAP1)-a membrane-bound mesenchymal stem cell marker of unknown function-as a highly expressed protein in Ewing tumors compared with benign tissues and show its regulation by EWS/FLI1. In addition, we show that STEAP1 knockdown reduces Ewing tumor proliferation, anchorage-independent colony formation as well as invasion in vitro and decreases growth and metastasis of Ewing tumor xenografts in vivo. Moreover, transcriptome and proteome analyses as well as functional studies revealed that STEAP1 expression correlates with oxidative stress responses and elevated levels of reactive oxygen species that in turn are able to regulate redox-sensitive and proinvasive genes. In synopsis, our data suggest that STEAP1 is associated with the invasive behavior and oxidative stress phenotype of Ewing tumors and point to a hitherto unanticipated oncogenic function of STEAP1.


Assuntos
Antígenos de Neoplasias/fisiologia , Neoplasias Ósseas/patologia , Estresse Oxidativo/genética , Oxirredutases/fisiologia , Sarcoma de Ewing/patologia , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Análise em Microsséries , Invasividade Neoplásica , Estresse Oxidativo/efeitos dos fármacos , Oxirredutases/antagonistas & inibidores , Oxirredutases/genética , Oxirredutases/metabolismo , Fenótipo , Proteômica , RNA Interferente Pequeno/farmacologia , Sarcoma de Ewing/genética , Sarcoma de Ewing/metabolismo
18.
Mol Biol Cell ; 22(22): 4424-34, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21965295

RESUMO

The vasoactive peptide urotensin-II (U-II) has been associated with vascular remodeling in different cardiovascular disorders. Although U-II can induce reactive oxygen species (ROS) by the NADPH oxidase NOX4 and stimulate smooth muscle cell (SMC) proliferation, the precise mechanisms linking U-II to vascular remodeling processes remain unclear. Forkhead Box O (FoxO) transcription factors have been associated with redox signaling and control of proliferation and apoptosis. We thus hypothesized that FoxOs are involved in the SMC response toward U-II and NOX4. We found that U-II and NOX4 stimulated FoxO activity and identified matrix metalloproteinase-2 (MMP2) as target gene of FoxO3a. FoxO3a activation by U-II was preceded by NOX4-dependent phosphorylation of c-Jun NH(2)-terminal kinase and 14-3-3 and decreased interaction of FoxO3a with its inhibitor 14-3-3, allowing MMP2 transcription. Functional studies in FoxO3a-depleted SMCs and in FoxO3a(-/-) mice showed that FoxO3a was important for basal and U-II-stimulated proliferation and vascular outgrowth, whereas treatment with an MMP2 inhibitor blocked these responses. Our study identified U-II and NOX4 as new activators of FoxO3a, and MMP2 as a novel target gene of FoxO3a, and showed that activation of FoxO3a by this pathway promotes vascular growth. FoxO3a may thus contribute to progression of cardiovascular diseases associated with vascular remodeling.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Músculo Liso Vascular/metabolismo , NADPH Oxidases/metabolismo , Urotensinas/metabolismo , Proteínas 14-3-3/metabolismo , Animais , Apoptose , Proliferação de Células , Células Cultivadas , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Metaloproteinase 2 da Matriz/biossíntese , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos Knockout , Músculo Liso Vascular/crescimento & desenvolvimento , Miócitos de Músculo Liso/metabolismo , NADPH Oxidase 4 , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
19.
Mol Biol Cell ; 21(12): 2087-96, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20427574

RESUMO

NADPH oxidases are important sources of reactive oxygen species (ROS), possibly contributing to various disorders associated with enhanced proliferation. NOX4 appears to be involved in vascular signaling and may contribute to the response to hypoxia. However, the exact mechanisms controlling NOX4 levels under hypoxia are not resolved. We found that hypoxia rapidly enhanced NOX4 mRNA and protein levels in pulmonary artery smooth-muscle cells (PASMCs) as well as in pulmonary vessels from mice exposed to hypoxia. This response was dependent on the hypoxia-inducible transcription factor HIF-1alpha because overexpression of HIF-1alpha increased NOX4 expression, whereas HIF-1alpha depletion prevented this response. Mutation of a putative hypoxia-responsive element in the NOX4 promoter abolished hypoxic and HIF-1alpha-induced activation of the NOX4 promoter. Chromatin immunoprecipitation confirmed HIF-1alpha binding to the NOX4 gene. Induction of NOX4 by HIF-1alpha contributed to maintain ROS levels after hypoxia and hypoxia-induced proliferation of PASMCs. These findings show that NOX4 is a new target gene of HIF-1alpha involved in the response to hypoxia. Together with our previous findings that NOX4 mediates HIF-1alpha induction under normoxia, these data suggest an important role of the signaling axis between NOX4 and HIF-1alpha in various cardiovascular disorders under hypoxic and also nonhypoxic conditions.


Assuntos
NADPH Oxidases/genética , Subunidades Proteicas/genética , Animais , Hipóxia Celular , Movimento Celular , Proliferação de Células , Indução Enzimática , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/enzimologia , NADPH Oxidase 4 , NADPH Oxidases/biossíntese , Subunidades Proteicas/biossíntese , Artéria Pulmonar/citologia , Espécies Reativas de Oxigênio/metabolismo , Transcrição Gênica
20.
Antioxid Redox Signal ; 13(4): 425-36, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20039838

RESUMO

The hypoxia-inducible factor-2alpha (HIF-2alpha) contributes to the vascular response to hypoxia. Hypoxia inhibits prolyl hydroxylation of the N-terminal transactivation domain (N-TAD), thus preventing binding of the von Hippel-Lindau protein (pVHL) and proteasomal degradation; additionally, hypoxia inhibits asparagyl hydroxylation of the C-TAD, thus diminishing cofactor recruitment. Reactive oxygen species (ROS) derived from NADPH oxidases (NOXs) have been shown to control vascular functions and to promote vascular remodeling. However, whether HIF-2alpha, ROS, and NOXs are linked under such nonhypoxic conditions is unclear. We found that activation of NOX4 by thrombin or H(2)O(2) increased HIF-2alpha protein because of decreased pVHL binding in pulmonary artery smooth muscle cells (PASMCs). Thrombin, H(2)O(2), and NOX4 overexpression increased HIF-2alpha N-TAD and C-TAD activity, which was prevented by ascorbate treatment or mutation of the hydroxylation sites in the TADs. HIF-2alpha also mediated induction of plasminogen activator inhibitor-1 and the proliferative response to thrombin, H(2)O(2), or NOX4 overexpression. Thus, ROS derived from NOX4 in response to thrombin stabilize HIF-2alpha by preventing hydroxylation of the N- and C-TAD, thus allowing formation of transcriptionally active HIF-2alpha, which promotes PASMC proliferation. Together, these findings present the first evidence that HIF-2alpha is critically involved in the ROS-regulated vascular remodeling processes.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , NADPH Oxidases/metabolismo , Estresse Oxidativo/fisiologia , Animais , Ácido Ascórbico/metabolismo , Ácido Ascórbico/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Humanos , Peróxido de Hidrogênio/farmacologia , Ferro/metabolismo , NADPH Oxidase 4 , NADPH Oxidases/genética , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Ratos , Espécies Reativas de Oxigênio/metabolismo , Trombina/farmacologia , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA