Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Int J Mol Sci ; 22(14)2021 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-34298974

RESUMO

CADASIL (Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy) is a small vessel disease caused by mutations in NOTCH3 that lead to an odd number of cysteines in the epidermal growth factor (EGF)-like repeat domain, causing protein misfolding and aggregation. The main symptoms are migraines, psychiatric disorders, recurrent strokes, and dementia. Omic technologies allow the massive study of different molecules for understanding diseases in a non-biased manner or even for discovering targets and their possible treatments. We analyzed the progress in understanding CADASIL that has been made possible by omics sciences. For this purpose, we included studies that focused on CADASIL and used omics techniques, searching bibliographic resources, such as PubMed. We excluded studies with other phenotypes, such as migraine or leukodystrophies. A total of 18 articles were reviewed. Due to the high prevalence of NOTCH3 mutations considered pathogenic to date in genomic repositories, one can ask whether all of them produce CADASIL, different degrees of the disease, or whether they are just a risk factor for small vessel disease. Besides, proteomics and transcriptomics studies found that the molecules that are significantly altered in CADASIL are mainly related to cell adhesion, the cytoskeleton or extracellular matrix components, misfolding control, autophagia, angiogenesis, or the transforming growth factor ß (TGFß) signaling pathway. The omics studies performed on CADASIL have been useful for understanding the biological mechanisms and could be key factors for finding potential drug targets.


Assuntos
CADASIL/fisiopatologia , Genômica/métodos , Proteômica/métodos , Receptor Notch3/genética , CADASIL/epidemiologia , CADASIL/genética , CADASIL/metabolismo , Cisteína/química , Microbioma Gastrointestinal , Frequência do Gene , Ontologia Genética , Estudos de Associação Genética , Estudo de Associação Genômica Ampla , Humanos , Modelos Moleculares , Mutação , Proteínas do Tecido Nervoso/análise , Prevalência , Prognóstico , Agregação Patológica de Proteínas/etiologia , Conformação Proteica , Domínios Proteicos , Receptor Notch3/química , Receptor Notch3/fisiologia , Análise de Sequência de DNA , Transcriptoma
2.
Sci Rep ; 10(1): 14839, 2020 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-32908186

RESUMO

Macrophage activation by Toll receptors is an essential event in the development of the response against pathogens. NOTCH signaling pathway is involved in the control of macrophage activation and the inflammatory processes. In this work, we have characterized NOTCH signaling in macrophages activated by Toll-like receptor (TLR) triggering and determined that DLL1 and DLL4 are the main ligands responsible for NOTCH signaling. We have identified ADAM10 as the main protease implicated in NOTCH processing and activation. We have also observed that furin, which processes NOTCH receptors, is induced by TLR signaling in a NOTCH-dependent manner. NOTCH3 is the only NOTCH receptor expressed in resting macrophages. Its expression increased rapidly in the first hours after TLR4 activation, followed by a gradual decrease, which was coincident with an elevation of the expression of the other NOTCH receptors. All NOTCH1, 2 and 3 contribute to the increased NOTCH signaling detected in activated macrophages. We also observed a crosstalk between NOTCH3 and NOTCH1 during macrophage activation. Finally, our results highlight the relevance of NOTCH3 in the activation of NF-κB, increasing p65 phosphorylation by p38 MAP kinase. Our data identify, for the first time, NOTCH3 as a relevant player in the control of inflammation.


Assuntos
Inflamação/imunologia , Macrófagos/imunologia , Receptor Notch3/fisiologia , Animais , Regulação da Expressão Gênica , Humanos , Ativação de Macrófagos , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , NF-kappa B/imunologia , Células RAW 264.7 , Transdução de Sinais , Receptores Toll-Like/imunologia
3.
Sci Rep ; 9(1): 18224, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31796853

RESUMO

Vascular smooth muscle cell (VSMC) dysfunction is a hallmark of small vessel disease, a common cause of stroke and dementia. Two of the most frequently mutated genes in familial small vessel disease are HTRA1 and NOTCH3. The protease HTRA1 cleaves the NOTCH3 ligand JAG1 implying a mechanistic link between HTRA1 and Notch signaling. Here we report that HTRA1 is essential for VSMC differentiation into the contractile phenotype. Mechanistically, loss of HTRA1 increased JAG1 protein levels and NOTCH3 signaling activity in VSMC. In addition, the loss of HTRA1 enhanced TGFß-SMAD2/3 signaling activity. Activation of either NOTCH3 or TGFß signaling resulted in increased transcription of the HES and HEY transcriptional repressors and promoted the contractile VSMC phenotype. However, their combined over-activation led to an additive accumulation of HES and HEY proteins, which repressed the expression of contractile VSMC marker genes. As a result, VSMC adopted an immature phenotype with impaired arterial vasoconstriction in Htra1-deficient mice. These data demonstrate an essential role of HTRA1 in vascular maturation and homeostasis by controlling Notch and TGFß signaling.


Assuntos
Serina Peptidase 1 de Requerimento de Alta Temperatura A/metabolismo , Músculo Liso Vascular/crescimento & desenvolvimento , Animais , Western Blotting , Imunofluorescência , Serina Peptidase 1 de Requerimento de Alta Temperatura A/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Muscular , Músculo Liso Vascular/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptor Notch3/metabolismo , Receptor Notch3/fisiologia , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/fisiologia
4.
Life Sci ; 232: 116582, 2019 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-31220525

RESUMO

AIMS: Vascular calcification/aging can cause different kind of serious diabetic vascular complications. High glucose could induce vascular smooth muscle cells (VSMCs) calcification/aging and then lead to diabetes-related vascular calcification/aging. In this study, we investigated how information in the blood is transmitted to VSMCs and the mechanisms of VSMCs calcification/aging under hyperglycaemic conditions. MATERIALS AND METHODS: Transmission electron microscopy and molecular size analysis were used to assess the morphology and size of exosomes. Alizarin Red S staining and senescence-associated ß galactosidase (SA-ß-gal) staining were carried out to detect calcification and senescence in VSMCs, respectively. Proteomics analysis was carried out to detect the different expression of exosomal proteins. Protein levels were measured by western blot analysis. KEY FINDINGS: The results show that exosomes isolated from high glucose stimulated human umbilical vein endothelial cell (HG-HUVEC-Exo) exhibited a bilayer structure morphology with a mean diameter of 63.63 ±â€¯2.96 nm. The presence of exosome markers including CD9, CD63 and TSG101 were also detected in HG-HUVEC-Exo. High glucose could induce VSMCs calcification/aging by increasing the expression of osteocalcin (OC) and p21 as well as the formation of mineralised nodules and SA-ß-gal positive cells. Fluorescence microscopy verified that the exosomes were taken up by VSMCs and Notch3 protein was enriched in HG-HUVEC-Exo. Most importantly, mTOR signalling was closely related to Notch3 protein and was involved in regulating HG-HUVEC-Exo-induced VSMCs calcification/aging. SIGNIFICANCE: The data demonstrate that Notch3 is required for HG-HUVEC-Exo promoted VSMCs calcification/aging and regulates VSMCs calcification/aging through the mTOR signalling pathway.


Assuntos
Músculo Liso Vascular/metabolismo , Receptor Notch3/fisiologia , Calcificação Vascular/metabolismo , Calcificação Fisiológica/efeitos dos fármacos , Cálcio/metabolismo , Células Cultivadas , Senescência Celular/fisiologia , Complicações do Diabetes/metabolismo , Complicações do Diabetes/fisiopatologia , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Exossomos/metabolismo , Glucose/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Hiperglicemia/metabolismo , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/metabolismo , Osteocalcina/metabolismo , Receptor Notch3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Calcificação Vascular/fisiopatologia
5.
Nat Commun ; 10(1): 1530, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30948783

RESUMO

Transition between differentiation states in development occurs swift but the mechanisms leading to epigenetic and transcriptional reprogramming are poorly understood. The pediatric cancer neuroblastoma includes adrenergic (ADRN) and mesenchymal (MES) tumor cell types, which differ in phenotype, super-enhancers (SEs) and core regulatory circuitries. These cell types can spontaneously interconvert, but the mechanism remains largely unknown. Here, we unravel how a NOTCH3 intracellular domain reprogrammed the ADRN transcriptional landscape towards a MES state. A transcriptional feed-forward circuitry of NOTCH-family transcription factors amplifies the NOTCH signaling levels, explaining the swift transition between two semi-stable cellular states. This transition induces genome-wide remodeling of the H3K27ac landscape and a switch from ADRN SEs to MES SEs. Once established, the NOTCH feed-forward loop maintains the induced MES state. In vivo reprogramming of ADRN cells shows that MES and ADRN cells are equally oncogenic. Our results elucidate a swift transdifferentiation between two semi-stable epigenetic cellular states.


Assuntos
Neurônios Adrenérgicos/patologia , Reprogramação Celular/genética , Células-Tronco Mesenquimais/patologia , Neuroblastoma/patologia , Receptor Notch3/fisiologia , Neurônios Adrenérgicos/metabolismo , Linhagem Celular Tumoral , Epigênese Genética , Retroalimentação Fisiológica , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Células-Tronco Mesenquimais/metabolismo , Neuroblastoma/metabolismo , Receptor Notch3/genética , Receptor Notch3/metabolismo
6.
Brain ; 142(4): 1009-1023, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30859180

RESUMO

We report a composite extreme phenotype design using distribution of white matter hyperintensities and brain infarcts in a population-based cohort of older persons for gene-mapping of cerebral small vessel disease. We demonstrate its application in the 3C-Dijon whole exome sequencing (WES) study (n = 1924, nWESextremes = 512), with both single variant and gene-based association tests. We used other population-based cohort studies participating in the CHARGE consortium for replication, using whole exome sequencing (nWES = 2,868, nWESextremes = 956) and genome-wide genotypes (nGW = 9924, nGWextremes = 3308). We restricted our study to candidate genes known to harbour mutations for Mendelian small vessel disease: NOTCH3, HTRA1, COL4A1, COL4A2 and TREX1. We identified significant associations of a common intronic variant in HTRA1, rs2293871 using single variant association testing (Pdiscovery = 8.21 × 10-5, Preplication = 5.25 × 10-3, Pcombined = 4.72 × 10-5) and of NOTCH3 using gene-based tests (Pdiscovery = 1.61 × 10-2, Preplication = 3.99 × 10-2, Pcombined = 5.31 × 10-3). Follow-up analysis identified significant association of rs2293871 with small vessel ischaemic stroke, and two blood expression quantitative trait loci of HTRA1 in linkage disequilibrium. Additionally, we identified two participants in the 3C-Dijon cohort (0.4%) carrying heterozygote genotypes at known pathogenic variants for familial small vessel disease within NOTCH3 and HTRA1. In conclusion, our proof-of-concept study provides strong evidence that using a novel composite MRI-derived phenotype for extremes of small vessel disease can facilitate the identification of genetic variants underlying small vessel disease, both common variants and those with rare and low frequency. The findings demonstrate shared mechanisms and a continuum between genes underlying Mendelian small vessel disease and those contributing to the common, multifactorial form of the disease.


Assuntos
Doenças de Pequenos Vasos Cerebrais/genética , Serina Peptidase 1 de Requerimento de Alta Temperatura A/genética , Receptor Notch3/genética , Idoso , Idoso de 80 Anos ou mais , Isquemia Encefálica/genética , Doenças de Pequenos Vasos Cerebrais/diagnóstico por imagem , Doenças de Pequenos Vasos Cerebrais/metabolismo , Estudos de Coortes , Feminino , Heterozigoto , Serina Peptidase 1 de Requerimento de Alta Temperatura A/metabolismo , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Mutação , Polimorfismo de Nucleotídeo Único , Receptor Notch3/metabolismo , Receptor Notch3/fisiologia , Acidente Vascular Cerebral/genética , Substância Branca/diagnóstico por imagem , Substância Branca/metabolismo , Sequenciamento do Exoma/métodos
7.
Genet Med ; 21(3): 676-682, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30032161

RESUMO

PURPOSE: CADASIL is a small-vessel disease caused by a cysteine-altering pathogenic variant in one of the 34 epidermal growth factor-like repeat (EGFr) domains of the NOTCH3 protein. We recently found that pathogenic variant in EGFr domains 7-34 have an unexpectedly high frequency in the general population (1:300). We hypothesized that EGFr 7-34 pathogenic variant more frequently cause a much milder phenotype, thereby explaining an important part of CADASIL disease variability. METHODS: Age at first stroke, survival and white matter hyperintensity volume were compared between 664 CADASIL patients with either a NOTCH3 EGFr 1-6 pathogenic variant or an EGFr 7-34 pathogenic variant. The frequencies of NOTCH3 EGFr 1-6 and EGFr 7-34 pathogenic variant were compared between individuals in the genome  Aggregation Database and CADASIL patients. RESULTS: CADASIL patients with an EGFr 1-6 pathogenic variant have a 12-year earlier onset of stroke than those with an EGFr 7-34 pathogenic variant, lower survival, and higher white matter hyperintensity volumes. Among diagnosed CADASIL patients, 70% have an EGFr 1-6 pathogenic variant, whereas EGFr 7-34 pathogenic variant strongly predominate in the population. CONCLUSION: NOTCH3 pathogenic variant position is the most important determinant of CADASIL disease severity, with EGFr 7-34 pathogenic variant predisposing to a later onset of stroke and longer survival.


Assuntos
CADASIL/genética , Receptor Notch3/genética , Adulto , Idoso , Encéfalo/patologia , CADASIL/fisiopatologia , Progressão da Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Países Baixos , Fenótipo , Domínios Proteicos/genética , Receptor Notch3/fisiologia , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/genética
8.
Proc Natl Acad Sci U S A ; 115(16): E3682-E3691, 2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29610298

RESUMO

Hemodynamic forces and Notch signaling are both known as key regulators of arterial remodeling and homeostasis. However, how these two factors integrate in vascular morphogenesis and homeostasis is unclear. Here, we combined experiments and modeling to evaluate the impact of the integration of mechanics and Notch signaling on vascular homeostasis. Vascular smooth muscle cells (VSMCs) were cyclically stretched on flexible membranes, as quantified via video tracking, demonstrating that the expression of Jagged1, Notch3, and target genes was down-regulated with strain. The data were incorporated in a computational framework of Notch signaling in the vascular wall, where the mechanical load was defined by the vascular geometry and blood pressure. Upon increasing wall thickness, the model predicted a switch-type behavior of the Notch signaling state with a steep transition of synthetic toward contractile VSMCs at a certain transition thickness. These thicknesses varied per investigated arterial location and were in good agreement with human anatomical data, thereby suggesting that the Notch response to hemodynamics plays an important role in the establishment of vascular homeostasis.


Assuntos
Proteína Jagged-1/fisiologia , Mecanotransdução Celular/fisiologia , Contração Muscular/fisiologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Receptor Notch3/fisiologia , Idoso , Artérias/ultraestrutura , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Simulação por Computador , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Homeostase , Humanos , Proteína Jagged-1/biossíntese , Proteína Jagged-1/genética , Ligantes , Pessoa de Meia-Idade , Modelos Biológicos , Morfogênese/fisiologia , Músculo Liso Vascular/ultraestrutura , Receptor Notch3/biossíntese , Receptor Notch3/genética , Proteínas Repressoras/biossíntese , Proteínas Repressoras/genética , Estresse Mecânico , Fatores de Transcrição HES-1/biossíntese , Fatores de Transcrição HES-1/genética , Gravação em Vídeo
9.
J Exp Med ; 214(8): 2271-2282, 2017 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-28698285

RESUMO

Cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is a neurological syndrome characterized by small vessel disease (SVD), stroke, and vascular cognitive impairment and dementia caused by mutations in NOTCH3 No therapies are available for this condition. Loss of mural cells, which encompass pericytes and vascular smooth muscle cells, is a hallmark of CADASIL and other SVDs, including diabetic retinopathy, resulting in vascular instability. Here, we showed that Notch3 signaling is both necessary and sufficient to support mural cell coverage in arteries using genetic rescue in Notch3 knockout mice. Furthermore, we show that systemic administration of an agonist Notch3 antibody prevents mural cell loss and modifies plasma proteins associated with Notch3 activity, including endostatin/collagen 18α1 and Notch3 extracellular domain in mice with the C455R mutation, a CADASIL variant associated with Notch3 loss of function. These findings open opportunities for the treatment of CADASIL and other SVDs by modulating Notch3 signaling.


Assuntos
Anticorpos/uso terapêutico , CADASIL/terapia , Receptor Notch3/fisiologia , Animais , Anticorpos/imunologia , Modelos Animais de Doenças , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiopatologia , Pericitos/fisiologia , Receptor Notch3/imunologia , Transdução de Sinais/fisiologia
10.
Zhonghua Gan Zang Bing Za Zhi ; 24(3): 202-6, 2016 Mar 20.
Artigo em Chinês | MEDLINE | ID: mdl-27095764

RESUMO

OBJECTIVE: To clarify whether HepG2 cells actively secrete Y-box binding protein-1 (YB-1) under stress conditions, and to investigate the pathological significance and mechanism of action of extracellular YB-1. METHODS: HepG2 cells were stimulated and treated by gradient concentrations of lipopolysaccharide (LPS) and adriamycin, the supernatant of the culture solution was collected by centrifugation, and the established chemiluminescence immunoassay (CLIA) was used for real-time quantitative determination of YB-1 level in the supernatant. The co-immunoprecipitation assay was used to detect whether extracellular YB-1 specifically bound to Notch3 receptor, and Western blot was used to measure the expression of Notch-NICD. The gradient concentrations of recombinant YB-1 were co-cultured with HepG2 cells, and MTT and migration assays were used to analyze the proliferation and invasion/metastasis of HepG2 cells. One-way analysis of variance was used for comparison of data between multiple groups. RESULTS: The results of CLIA confirmed that the level of extracellular YB-1 in the supernatant was significantly higher than that in the control group (F= 10.54,P< 0.001), and the secretory expression of YB-1 reached its peak after 4 hours of stimulation (LPS: 8 ng/ml; adriamycin: 10 ng/ml). The results of co-immunoprecipitation assay and Western blot showed that extracellular YB-1 specifically bound to Notch3 receptor and upregulated the expression of the Notch3 receptor. MTT and migration assays showed that extracellular YB-1 significantly promoted the proliferation and invasion/metastasis of HepG2 cells (F= 9.405,P< 0.001). CONCLUSION: Under the stress conditions induced by chemotherapeutics, HepG2 cells can actively secrete YB-1 via non-classical pathways. Extracellular YB-1 can specifically bind to Notch3 receptor and further up-regulate its expression, and then promote the proliferation and invasion/metastasis of HepG2 cells. This study lays a foundation for further clarifying the pathogenesis of hepatocellular carcinoma and investigating the biological relationship between extracellular YB-1 and malignant tumors.


Assuntos
Carcinoma Hepatocelular/patologia , Células Hep G2 , Neoplasias Hepáticas/patologia , Receptor Notch3 , Proteína 1 de Ligação a Y-Box , Carcinoma Hepatocelular/metabolismo , Humanos , Lipopolissacarídeos , Neoplasias Hepáticas/metabolismo , Receptor Notch3/fisiologia , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA