Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 128(3): 401-418, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33322916

RESUMEN

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.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Células Endoteliales/efectos de los fármacos , Imidazoles/farmacología , Neovascularización Fisiológica/efectos de los fármacos , PPAR gamma/metabolismo , Piperazinas/farmacología , Hipertensión Arterial Pulmonar/tratamiento farmacológico , Arteria Pulmonar/efectos de los fármacos , Regeneración/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Células Cultivadas , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , Estrés Oxidativo , PPAR gamma/genética , Hipertensión Arterial Pulmonar/genética , Hipertensión Arterial Pulmonar/metabolismo , Hipertensión Arterial Pulmonar/fisiopatología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Transducción de Señal , Proteína p53 Supresora de Tumor/genética
2.
BMC Neurol ; 22(1): 469, 2022 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-36494773

RESUMEN

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.


Asunto(s)
Neuropatías Amiloides Familiares , Cardiomiopatías , Humanos , Neuropatías Amiloides Familiares/complicaciones , Neuropatías Amiloides Familiares/diagnóstico , Neuropatías Amiloides Familiares/genética , Mutación/genética , Cardiomiopatías/diagnóstico , Cardiomiopatías/genética , Fenotipo , Progresión de la Enfermedad
3.
Hum Mutat ; 41(5): 1025-1032, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32048431

RESUMEN

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.


Asunto(s)
Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Alelos , Bases de Datos Genéticas , Femenino , Estudios de Asociación Genética/métodos , Pruebas Genéticas , Variación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Fenotipo , Síndrome
4.
Mol Cell Probes ; 44: 14-20, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30682426

RESUMEN

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.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Subunidad alfa de la Proteína Trifuncional Mitocondrial/genética , Subunidad beta de la Proteína Trifuncional Mitocondrial/genética , Mutación/genética , Adolescente , Cardiomiopatías/genética , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Lactante , Errores Innatos del Metabolismo Lipídico/genética , Masculino , Miopatías Mitocondriales/genética , Proteína Trifuncional Mitocondrial/deficiencia , Proteína Trifuncional Mitocondrial/genética , Enfermedades del Sistema Nervioso/genética , Fenotipo , Rabdomiólisis/genética , Síndrome
5.
Circulation ; 133(18): 1783-94, 2016 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-27045138

RESUMEN

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.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/deficiencia , Transición Epitelial-Mesenquimal/fisiología , Proteína HMGA1a/biosíntesis , Hipertensión Pulmonar/metabolismo , Factores de Transcripción de la Familia Snail/biosíntesis , Adolescente , Adulto , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Células Cultivadas , Niño , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Proteína HMGA1a/genética , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Lactante , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Factores de Transcripción de la Familia Snail/genética , Adulto Joven
6.
Am J Respir Crit Care Med ; 191(11): 1273-86, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25853696

RESUMEN

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.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/efectos de los fármacos , Caveolina 1/efectos de los fármacos , Elafina/farmacología , Hipertensión Pulmonar/tratamiento farmacológico , Inhibidores de Proteasas/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Humanos , Miocitos del Músculo Liso/efectos de los fármacos , Elastasa Pancreática/efectos de los fármacos , Ratas
7.
J Cell Sci ; 125(Pt 4): 956-64, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22399808

RESUMEN

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.


Asunto(s)
Factor 1 Inducible por Hipoxia/metabolismo , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasas/metabolismo , Neovascularización Fisiológica , Urotensinas/metabolismo , Animales , Retroalimentación Fisiológica , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , NADPH Oxidasa 2 , NADPH Oxidasas/biosíntesis , NADPH Oxidasas/deficiencia , NADPH Oxidasas/genética , Neovascularización Patológica , Neovascularización Fisiológica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Urotensinas/farmacología
9.
Circ Res ; 104(10): 1169-77, 2009 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-19390057

RESUMEN

Cyclic nucleotide phosphodiesterases (PDEs) control the levels of the second messengers cAMP and cGMP in many cell types including endothelial cells. Although PDE2 has the unique property to be activated by cGMP but to hydrolyze cAMP, its role in endothelial function is only poorly understood. Reactive oxygen species (ROS) have been recognized as signaling molecules controlling many endothelial functions. We thus investigated whether PDE2 would link to ROS generation and proliferative responses in human umbilical vein endothelial cells in response to thrombin. Thrombin stimulated the GTPase Rac1, known to activate NADPH oxidases, and enhanced ROS formation, whereas PDE2 inhibition or depletion by short hairpin (sh)RNA prevented these responses. Similar observations were made with 8-Br-cGMP or atrial natriuretic peptide. In agreement, thrombin elevated cGMP but decreased cAMP levels, whereas db-cAMP or forskolin diminished Rac1 activity and ROS production. Subsequently, PDE2 overexpression activated Rac1, increased ROS generation, and enhanced proliferation and in vitro capillary formation. These responses were not observed in the presence of inactive Rac1 or shRNA against the NADPH oxidase subunit NOX2. Inhibition or depletion of PDE2 also prevented thrombin-induced proliferation and capillary formation. Importantly, downregulation of PDE2 by lentiviral shRNA or PDE2 inhibition prevented vessel sprouting from mouse aortic explants and in vivo angiogenesis in a mouse model, respectively. In summary, PDE2 promotes activation of NADPH oxidase-dependent ROS production and subsequent endothelial proliferation and angiogenesis. Targeting PDE2 may provide a new therapeutic approach in diseases associated with endothelial dysfunction, oxidative stress, vascular proliferation, and angiogenesis.


Asunto(s)
Proliferación Celular , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Endotelio Vascular/metabolismo , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasas/metabolismo , Neovascularización Fisiológica/fisiología , Trombina/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Aorta/citología , Aorta/metabolismo , Células Cultivadas , GMP Cíclico/metabolismo , Endotelio Vascular/citología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasa 2 , Especies Reactivas de Oxígeno/metabolismo , Venas Umbilicales/citología , Venas Umbilicales/metabolismo , Quinasas p21 Activadas/metabolismo
10.
Cardiovasc Diagn Ther ; 11(2): 637-649, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33968641

RESUMEN

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.

11.
BMC Med Genomics ; 14(1): 94, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33789662

RESUMEN

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.


Asunto(s)
Secuenciación del Exoma , Autopsia , Biología Computacional , Exoma , Humanos , Masculino , Fenotipo
12.
Neuromuscul Disord ; 31(2): 123-133, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33414056

RESUMEN

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.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Estudios de Asociación Genética , Mutación/genética , Ubiquitina-Proteína Ligasas , Adolescente , Adulto , Anciano , Axones/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Ubiquitina-Proteína Ligasas/genética , Adulto Joven
13.
Parkinsonism Relat Disord ; 90: 73-78, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34399161

RESUMEN

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.


Asunto(s)
Encefalopatías/genética , Distonía/genética , Síndromes Epilépticos/genética , Trastornos del Neurodesarrollo/genética , Adolescente , Encefalopatías/complicaciones , Niño , Preescolar , Síndromes Epilépticos/complicaciones , Femenino , Factores de Transcripción Forkhead/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Humanos , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Proteínas del Tejido Nervioso/genética , Trastornos del Neurodesarrollo/complicaciones , Fenotipo , Receptores de GABA-A/genética , Receptores de GABA-B/genética
14.
Cell Rep ; 26(5): 1333-1343.e7, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30699358

RESUMEN

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.


Asunto(s)
Reparación del ADN , Células Endoteliales/metabolismo , Homeostasis , PPAR gamma/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Daño del ADN , Inestabilidad Genómica , Células HEK293 , Humanos , Modelos Biológicos , Unión Proteica , Arteria Pulmonar/patología , Transducción de Señal , Ubiquitinación
15.
Thromb Haemost ; 100(6): 984-91, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19132221

RESUMEN

Vascular remodelling is a complex phenomenon associated with restructuring of the vessel wall as a consequence of disruption of vascular homeostasis. Alterations of the vascular wall have been linked to a variety of cardiovascular disorders including atherosclerosis, vascular injury and pulmonary hypertension. Plasminogen activator inhibitor-1 (PAI-1) is a member of the serpin (serine proteinase inhibitor) family and acts as an important inhibitor of fibrinolysis by interfering with the plasminogen system. In addition to its anti-fibrinolytic effects, PAI-1 appears to modulate cellular responses linked to vascular remodelling. Since PAI-1 levels have been shown to be altered in various disorders associated with vascular remodelling of the systemic and pulmonary vascular bed, this serpin may play a pivotal role in the pathogenesis of these diseases.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Endotelio Vascular/metabolismo , Músculo Liso Vascular/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Transducción de Señal , Animales , Aterosclerosis/metabolismo , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/patología , Proliferación Celular , Endotelio Vascular/lesiones , Endotelio Vascular/patología , Humanos , Hipertensión Pulmonar , Ratones , Ratones Noqueados , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , Inhibidor 1 de Activador Plasminogénico/genética , Arteria Pulmonar/metabolismo
16.
Thromb Haemost ; 100(6): 1021-8, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19132225

RESUMEN

Pulmonary vascular remodeling is commonly associated with pulmonary hypertension and is characterized by media thickening and disordered cellular proliferation, often accompanied by fibrin deposition and thrombosis in situ. However, the signaling pathways linking these different processes are not well understood. Since the GTPase Rac-1 has been suggested to act as a signaling relay in various cell types we investigated whether Rac-1 could be the link between thrombin signaling, plasminogen activator inhibitor-1 (PAI-1), which inhibits fibrinolysis and promotes fibrin deposition, and proliferation of pulmonary artery smooth muscle cells (PASMC). Exposure to thrombin enhanced the levels of Rac-1 protein and increased PAI-1 mRNA and protein expression in dependence of the thrombin receptor PAR-1. Expression of dominant-negative Rac-1 (RacT17N) prevented thrombin-induced PAI-1 expression whereas constitutively active RacG12V enhanced PAI-1 levels. In the presence of RacT17N thrombin-induced PAI-1 promoter activity was abrogated whereas RacG12V increased PAI-1 promoter activity, and this response was essentially dependent on the transcription factor hypoxia-inducible factor-1 (HIF-1). Subsequently, RacG12V not only increased HIF transcriptional activity but also HIF-1alpha protein and mRNA levels, whereas RacT17N prevented these responses elicited by thrombin. In line, RacG12V enhanced HIF-1alpha promoter activity, and this response was dependent on nuclear factor-kappaB (NFkappaB) binding to the HIF-1alpha promoter. Finally, upregulation of PAI-1 by Rac-1 and HIF-1 was essential for thrombin-stimulated proliferation of PASMC. These findings indicate that Rac-1 is an important mediator of thrombin signaling and may contribute to pulmonary vascular remodeling via HIF-1-dependent upregulation of PAI-1 leading to enhanced proliferation of PASMC.


Asunto(s)
Proliferación Celular , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , FN-kappa B/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Transcripción Genética , Proteína de Unión al GTP rac1/metabolismo , Animales , Células Cultivadas , Humanos , Inhibidor 1 de Activador Plasminogénico/genética , Arteria Pulmonar/enzimología , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Receptor PAR-1/metabolismo , Transducción de Señal , Trombina/metabolismo , Factores de Tiempo , Transfección , Regulación hacia Arriba
17.
Mol Genet Genomic Med ; 6(6): 1188-1198, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30406974

RESUMEN

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.


Asunto(s)
Pruebas Genéticas/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Enfermedades Mitocondriales/genética , Análisis de Secuencia de ADN/métodos , Costos y Análisis de Costo , ADN Mitocondrial/química , ADN Mitocondrial/genética , Pruebas Genéticas/economía , Pruebas Genéticas/normas , Secuenciación de Nucleótidos de Alto Rendimiento/economía , Secuenciación de Nucleótidos de Alto Rendimiento/normas , Humanos , Enfermedades Mitocondriales/diagnóstico , Sensibilidad y Especificidad , Análisis de Secuencia de ADN/economía , Análisis de Secuencia de ADN/normas
18.
Sci Rep ; 8(1): 11682, 2018 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-30076399

RESUMEN

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.


Asunto(s)
ADN Mitocondrial/genética , Células Musculares/metabolismo , Reacción en Cadena de la Polimerasa/métodos , Eliminación de Secuencia/genética , Adolescente , Adulto , Anciano , Biopsia , Complejo IV de Transporte de Electrones/metabolismo , Femenino , GTP Fosfohidrolasas/genética , Dosificación de Gen , Genes Recesivos , Humanos , Masculino , Persona de Mediana Edad , Fibras Musculares Esqueléticas/metabolismo , Mutación/genética , Fosforilación Oxidativa , Reproducibilidad de los Resultados , Succinato Deshidrogenasa/metabolismo , Adulto Joven
19.
Circulation ; 105(17): 2030-6, 2002 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-11980681

RESUMEN

BACKGROUND: Tissue factor (TF) initiates the extrinsic coagulation cascade leading to thrombin formation. Thrombin induces TF mRNA in vascular smooth muscle cells (VSMCs), thereby contributing to the prolonged procoagulant activity and enhanced thrombogenicity at sites of vascular injury. However, the signaling mechanisms mediating this thrombogenic cycle are unclear. Characteristically, vascular injury promotes the generation of reactive oxygen species (ROS). Because ROS exert signaling functions, we investigated whether the NADPH oxidase, an important source of ROS in VSMCs, contributes to upregulation of TF by thrombin. METHODS AND RESULTS: Thrombin not only stimulated TF mRNA expression, but also TF-dependent surface procoagulant activity in cultured human VSMCs. This response was attenuated by antioxidants; the flavin inhibitor diphenylene-iodonium, Clostridium difficile toxin B, which inhibits Rho GTPases, p22phox antisense oligonucleotides, or the dominant-negative RacT17N mutant. Inhibitors of p38 MAP kinase and phosphatidylinositol (PI) 3-kinase also prevented thrombin-stimulated TF mRNA expression. Furthermore, thrombin stimulated the phosphorylation of the PI 3-kinase target protein kinase B/Akt in a redox-sensitive and NADPH oxidase-dependent manner. CONCLUSION: These findings indicate that the NADPH oxidase is essentially involved in the redox-sensitive induction of TF mRNA expression and surface procoagulant activity by thrombin. This response is mediated by NADPH oxidase-dependent activation of p38 MAP kinase and the PI 3-kinase/protein kinase B/Akt pathway. Given that active TF promotes thrombin formation, the NADPH oxidase may play a crucial role in perpetuating the thrombogenic cycle in the injured vessel wall.


Asunto(s)
Proteínas Bacterianas , Coagulantes/farmacología , Proteínas de Transporte de Membrana , Músculo Liso Vascular/metabolismo , NADPH Oxidasas/fisiología , Proteínas Serina-Treonina Quinasas , Trombina/farmacología , Tromboplastina/metabolismo , Toxinas Bacterianas/farmacología , Membrana Celular/metabolismo , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Humanos , Cinética , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/enzimología , NADPH Deshidrogenasa/genética , NADPH Oxidasas/antagonistas & inhibidores , Oligonucleótidos Antisentido/farmacología , Fosfoproteínas/genética , Inhibidores de Proteínas Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , ARN Mensajero/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Tromboplastina/genética , Activación Transcripcional/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rho/antagonistas & inhibidores
20.
Cell Metab ; 21(4): 596-608, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25863249

RESUMEN

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.


Asunto(s)
Supervivencia Celular/fisiología , Células Endoteliales/fisiología , Hipertensión Pulmonar/metabolismo , Mitocondrias/metabolismo , Modelos Biológicos , Arteria Pulmonar/fisiología , Regeneración/fisiología , Análisis de Varianza , Animales , Western Blotting , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , ADN/metabolismo , Cartilla de ADN/genética , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Hipertensión Pulmonar/fisiopatología , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Reacción en Cadena de la Polimerasa , Arteria Pulmonar/citología , ARN Interferente Pequeño/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA