Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nature ; 604(7907): 740-748, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35444273

RESUMEN

All tissue-resident macrophages of the central nervous system (CNS)-including parenchymal microglia, as well as CNS-associated macrophages (CAMs1) such as meningeal and perivascular macrophages2-7-are part of the CNS endogenous innate immune system that acts as the first line of defence during infections or trauma2,8-10. It has been suggested that microglia and all subsets of CAMs are derived from prenatal cellular sources in the yolk sac that were defined as early erythromyeloid progenitors11-15. However, the precise ontogenetic relationships, the underlying transcriptional programs and the molecular signals that drive the development of distinct CAM subsets in situ are poorly understood. Here we show, using fate-mapping systems, single-cell profiling and cell-specific mutants, that only meningeal macrophages and microglia share a common prenatal progenitor. By contrast, perivascular macrophages originate from perinatal meningeal macrophages only after birth in an integrin-dependent manner. The establishment of perivascular macrophages critically requires the presence of arterial vascular smooth muscle cells. Together, our data reveal a precisely timed process in distinct anatomical niches for the establishment of macrophage subsets in the CNS.


Asunto(s)
Linaje de la Célula , Sistema Nervioso Central , Macrófagos , Sistema Nervioso Central/inmunología , Femenino , Humanos , Inmunidad Innata , Macrófagos/citología , Microglía , Embarazo , Saco Vitelino
2.
Elife ; 52016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27476853

RESUMEN

Cerebral small vessel disease (SVD) is a leading cause of stroke and dementia. CADASIL, an inherited SVD, alters cerebral artery function, compromising blood flow to the working brain. TIMP3 (tissue inhibitor of metalloproteinase 3) accumulation in the vascular extracellular matrix in CADASIL is a key contributor to cerebrovascular dysfunction. However, the linkage between elevated TIMP3 and compromised cerebral blood flow (CBF) remains unknown. Here, we show that TIMP3 acts through inhibition of the metalloprotease ADAM17 and HB-EGF to regulate cerebral arterial tone and blood flow responses. In a clinically relevant CADASIL mouse model, we show that exogenous ADAM17 or HB-EGF restores cerebral arterial tone and blood flow responses, and identify upregulated voltage-dependent potassium channel (KV) number in cerebral arterial myocytes as a heretofore-unrecognized downstream effector of TIMP3-induced deficits. These results support the concept that the balance of TIMP3 and ADAM17 activity modulates CBF through regulation of myocyte KV channel number.


Asunto(s)
Proteína ADAM17/metabolismo , Encéfalo/fisiología , CADASIL/fisiopatología , Hemodinámica , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Canales de Potasio con Entrada de Voltaje/metabolismo
3.
Brain Pathol ; 24(5): 525-44, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25323668

RESUMEN

CADASIL and CARASIL are hereditary small vessel diseases leading to vascular dementia. CADASIL commonly begins with migraine followed by minor strokes in mid-adulthood. Dominantly inherited CADASIL is caused by mutations (n > 230) in NOTCH3 gene, which encodes Notch3 receptor expressed in vascular smooth muscle cells (VSMC). Notch3 extracellular domain (N3ECD) accumulates in arterial walls followed by VSMC degeneration and subsequent fibrosis and stenosis of arterioles, predominantly in cerebral white matter, where characteristic ischemic MRI changes and lacunar infarcts emerge. The likely pathogenesis of CADASIL is toxic gain of function related to mutation-induced unpaired cysteine in N3ECD. Definite diagnosis is made by molecular genetics but is also possible by electron microscopic demonstration of pathognomonic granular osmiophilic material at VSMCs or by positive immunohistochemistry for N3ECD in dermal arteries. In rare, recessively inherited CARASIL the clinical picture and white matter changes are similar as in CADASIL, but cognitive decline begins earlier. In addition, gait disturbance, low back pain and alopecia are characteristic features. CARASIL is caused by mutations (presently n = 10) in high-temperature requirement. A serine peptidase 1 (HTRA1) gene, which result in reduced function of HTRA1 as repressor of transforming growth factor-ß (TGF ß) -signaling. Cerebral arteries show loss of VSMCs and marked hyalinosis, but not stenosis.


Asunto(s)
Alopecia/genética , Alopecia/patología , CADASIL/genética , CADASIL/patología , Infarto Cerebral/genética , Infarto Cerebral/patología , Leucoencefalopatías/genética , Leucoencefalopatías/patología , Enfermedades de la Columna Vertebral/genética , Enfermedades de la Columna Vertebral/patología , Encéfalo/irrigación sanguínea , Encéfalo/patología , Humanos
4.
Stroke ; 45(3): 842-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24425116

RESUMEN

BACKGROUND AND PURPOSE: Cerebral autosomal dominant arteriopathy with subcortical infarcts and leucoencephalopathy, the most common heritable small vessel disease of the brain, is caused by dominant mutations in the NOTCH3 receptor that stereotypically lead to age-dependent Notch3ECD deposition in the vessels. NOTCH3 loss of function has been demonstrated for few mutations. However, whether this finding applies to all mutations and whether a loss-of-function mechanism drives the manifestations of the disease remain yet unknown. This study investigated the in vivo functionality of the Arg169Cys archetypal mutation. METHODS: We used mice with constitutive or conditional reduction of NOTCH3 activity, mice harboring the Arg169Cys mutation at the endogenous Notch3 locus (Notch3Arg170Cys), and mice overexpressing the Arg169Cys NOTCH3 mutant (TgPAC-Notch3R169C) on either a Notch3 wild-type or a null background. NOTCH3 activity was monitored in the brain arteries by measuring the expression of NOTCH3 target genes using real-time polymerase chain reaction. Notch3ECD deposits were assessed by immunohistochemistry. Brain parenchyma was analyzed for vacuolation and myelin debris in the white matter and infarcts. RESULTS: We identified a subset of genes appropriate to detect NOTCH3 haploinsufficiency in the adult. Expression of these genes was unaltered in Notch3Arg170Cys mice, despite marked Notch3ECD deposits. Elimination of wild-type NOTCH3 did not influence the onset and burden of white matter lesions in 20-month-old TgPAC-Notch3R169C mice, and 20-month-old Notch3-null mice exhibited neither infarct nor white matter changes. CONCLUSIONS: These data provide strong evidence that cerebral autosomal dominant arteriopathy with subcortical infarcts and leucoencephalopathy can develop without impairment of NOTCH3 signaling and argue against a loss of NOTCH3 function as a general driving mechanism for white matter lesions in cerebral autosomal dominant arteriopathy with subcortical infarcts and leucoencephalopathy.


Asunto(s)
CADASIL/genética , Mutación/genética , Mutación/fisiología , Receptores Notch/genética , Receptores Notch/fisiología , Animales , Encéfalo/patología , CADASIL/patología , Arterias Cerebrales/patología , Antagonistas de Estrógenos/farmacología , Regulación de la Expresión Génica , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Notch3 , Tamoxifeno/farmacología , Transcripción Genética
5.
Brain ; 136(Pt 6): 1830-45, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23649698

RESUMEN

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy, or CADASIL, one of the most common inherited small vessel diseases of the brain, is characterized by a progressive loss of vascular smooth muscle cells and extracellular matrix accumulation. The disease is caused by highly stereotyped mutations within the extracellular domain of the NOTCH3 receptor (Notch3(ECD)) that result in an odd number of cysteine residues. While CADASIL-associated NOTCH3 mutations differentially affect NOTCH3 receptor function and activity, they all are associated with early accumulation of Notch3(ECD)-containing aggregates in small vessels. We still lack mechanistic explanation to link NOTCH3 mutations with small vessel pathology. Herein, we hypothesized that excess Notch3(ECD) could recruit and sequester functionally important proteins within small vessels of the brain. We performed biochemical, nano-liquid chromatography-tandem mass spectrometry and immunohistochemical analyses, using cerebral and arterial tissue derived from patients with CADASIL and mouse models of CADASIL that exhibit vascular lesions in the end- and early-stage of the disease, respectively. Biochemical fractionation of brain and artery samples demonstrated that mutant Notch3(ECD) accumulates in disulphide cross-linked detergent-insoluble aggregates in mice and patients with CADASIL. Further proteomic and immunohistochemical analyses identified two functionally important extracellular matrix proteins, tissue inhibitor of metalloproteinases 3 (TIMP3) and vitronectin (VTN) that are sequestered into Notch3(ECD)-containing aggregates. Using cultured cells, we show that increased levels or aggregation of Notch3 enhances the formation of Notch3(ECD)-TIMP3 complex, promoting TIMP3 recruitment and accumulation. In turn, TIMP3 promotes complex formation including NOTCH3 and VTN. In vivo, brain vessels from mice and patients with CADASIL exhibit elevated levels of both insoluble cross-linked and soluble TIMP3 species. Moreover, reverse zymography assays show a significant elevation of TIMP3 activity in the brain vessels from mice and patients with CADASIL. Collectively, our findings lend support to a Notch3(ECD) cascade hypothesis in CADASIL disease pathology, which posits that aggregation/accumulation of Notch3(ECD) in the brain vessels is a central event, promoting the abnormal recruitment of functionally important extracellular matrix proteins that may ultimately cause multifactorial toxicity. Specifically, our results suggest a dysregulation of TIMP3 activity, which could contribute to mutant Notch3(ECD) toxicity by impairing extracellular matrix homeostasis in small vessels.


Asunto(s)
CADASIL/diagnóstico , CADASIL/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Receptores Notch/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Anciano , Anciano de 80 o más Años , Animales , CADASIL/genética , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/genética , Femenino , Homeostasis/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Transporte de Proteínas/genética , Receptor Notch3 , Receptores Notch/genética , Inhibidor Tisular de Metaloproteinasa-3/genética
6.
Bioessays ; 33(1): 73-80, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20967782

RESUMEN

Small vessel diseases (SVDs) of the brain are the leading cause of vascular cognitive impairment and a major contributor to stroke in the human adult, however, their pathogenesis is poorly understood. Dominant mutations in NOTCH3 cause CADASIL, one of the most prevalent inherited cerebral SVDs. The disease gene encodes a transmembrane receptor primarily expressed in smooth muscle cells of small arteries and pericytes of brain capillaries. Pathogenic mutations alter the number of cysteine residues in the extracellular domain of NOTCH3, leading to its abnormal accumulation in the vessels of patients. Mice lacking NOTCH3 have revealed a critical role for NOTCH3 in the elaboration of small arteries. Despite being incomplete disease models, transgenic mice expressing CADASIL-associated NOTCH3 mutations, have provided important insights into specific aspects of CADASIL pathogenesis, including the functional significance of disease-linked mutations and the earliest pathological events that initiate brain lesions. In this paper, I provide a critical overview of these studies. Moreover, I discuss future directions and further work that needs to be done.


Asunto(s)
Arterias/metabolismo , CADASIL , Receptores Notch , Adulto , Animales , Arterias/patología , Arterias/fisiopatología , Encéfalo/patología , Encéfalo/fisiología , Encéfalo/fisiopatología , CADASIL/genética , CADASIL/patología , CADASIL/fisiopatología , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación/fisiología , Receptor Notch3 , Receptores Notch/genética , Receptores Notch/metabolismo
7.
Lancet Neurol ; 8(7): 643-53, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19539236

RESUMEN

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leucoencephalopathy (CADASIL) is the most common heritable cause of stroke and vascular dementia in adults. Clinical and neuroimaging features resemble those of sporadic small-artery disease, although patients with CADASIL have an earlier age at onset of stroke events, an increased frequency of migraine with aura, and a slightly variable pattern of ischaemic white-matter lesions on brain MRI. NOTCH3 (Notch homolog 3), the gene involved in CADASIL, encodes a transmembrane receptor primarily expressed in systemic arterial smooth-muscle cells. Pathogenetic mutations alter the number of cysteine residues in the extracellular domain of NOTCH3, which accumulates in small arteries of affected individuals. Functional and imaging studies in cultured cells, genetically engineered mice, and patients with CADASIL have all provided insights into the molecular and vascular mechanisms underlying this disease. A recent multicentre trial in patients with cognitive impairment emphasises the feasibility of randomised trials in patients with CADASIL. In this Review, we summarise the current understanding of CADASIL, a devastating disorder that also serves as a model for the more common forms of subcortical ischaemic strokes and pure vascular dementia.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/patología , CADASIL/patología , Arterias Cerebrales/patología , Arterias Cerebrales/fisiopatología , Infarto Encefálico/etiología , Infarto Encefálico/patología , Infarto Encefálico/fisiopatología , Isquemia Encefálica/etiología , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , CADASIL/genética , CADASIL/terapia , Arterias Cerebrales/metabolismo , Progresión de la Enfermedad , Predisposición Genética a la Enfermedad/genética , Humanos , Migraña con Aura/etiología , Migraña con Aura/patología , Migraña con Aura/fisiopatología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiopatología , Receptor Notch3 , Receptores Notch/genética
8.
Brain ; 132(Pt 6): 1601-12, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19293235

RESUMEN

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is an autosomal dominant small-vessel disease of the brain caused by mutations in the NOTCH3 receptor. The highly stereotyped nature of the mutations, which alter the number of cysteine residues within the epidermal growth factor-like repeats (EGFR), predicts that all mutations share common mechanisms. Prior in vitro assays and genetic studies in the mouse support the hypothesis that common mutations do not compromise canonical Notch3 function but instead convey a non-physiological and deleterious activity to the receptor through the unpaired cysteine residue. Intriguingly, in vitro studies predict that mutations located in the Delta/Serrate/LAG-2 ligand binding domain-(EGFR10-11) may result in a loss of Notch3 receptor function. However, the in vivo relevance and functional significance of this with respect to the pathogenic mechanisms and clinical expression of the disease remain largely unexplored. To ascertain, in vivo, the functional significance of EGFR10-11 mutations, we generated transgenic mice with one representative mutation (C428S) in EGFR10 of Notch3. These mice, like those with a common R90C mutation, developed characteristic arterial accumulation of Notch3 protein and granular osmiophilic material upon aging. By introducing the mutant C428S transgene into a Notch3 null background, we found that, unlike the R90C mutant protein, the C428S mutant protein has lost wild-type Notch3 activity and exhibited mild dominant-negative activity in three different biological settings. From a large prospectively recruited cohort of 176 CADASIL patients, we identified 10 patients, from five distinct pedigrees carrying a mutation in EGFR10 or 11. These mutations were associated with significantly higher Mini-Mental State Examination and Mattis Dementia Rating Scale scores (P < 0.05), when compared with common mutations. Additionally, we found a strong effect of this genotype on the burden of white matter hyperintensities (P < 0.01). Collectively, these results highlight distinctive functional and phenotypic features of EGFR10-11 mutations relative to the common CADASIL mutations. Our findings are compatible with the hypothesis that EGFR10-11 mutations cause the disease through the same gain of novel function as the common mutations, and lead us to propose that reduced Notch3 signalling acts as a modifier of the CADASIL phenotype.


Asunto(s)
CADASIL/genética , Mutación , Receptores Notch/genética , Adulto , Anciano , Animales , Encéfalo/patología , CADASIL/metabolismo , CADASIL/patología , Arterias Cerebrales/metabolismo , Arterias Cerebrales/ultraestructura , Modelos Animales de Enfermedad , Genotipo , Humanos , Ligandos , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Transgénicos , Microscopía Electrónica , Persona de Mediana Edad , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/ultraestructura , Fenotipo , Estudios Prospectivos , Receptor Notch3 , Receptores Notch/metabolismo , Receptores Notch/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
9.
Brain ; 132(Pt 4): 933-9, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19174371

RESUMEN

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common hereditary subcortical vascular dementia. It is caused by mutations in NOTCH3 gene, which encodes a large transmembrane receptor Notch3. The key pathological finding is the accumulation of granular osmiophilic material (GOM), which contains extracellular domains of Notch3, on degenerating vascular smooth muscle cells (VSMCs). GOM has been considered specifically diagnostic for CADASIL, but the reports on the sensitivity of detecting GOM in patients' skin biopsy have been contradictory. To solve this contradiction, we performed a retrospective investigation of 131 Finnish, Swedish and French CADASIL patients, who had been adequately examined for both NOTCH3 mutation and presence of GOM. The patients were examined according to the diagnostic practice in each country. NOTCH3 mutations were assessed by restriction enzyme analysis of specific mutations or by sequence analysis. Presence of GOM was examined by electron microscopy (EM) in skin biopsies. Biopsies of 26 mutation-negative relatives from CADASIL families served as the controls. GOM was detected in all 131 mutation positive patients. Altogether our patients had 34 different pathogenic mutations which included three novel point mutations (p.Cys67Ser, p.Cys251Tyr and p.Tyr1069Cys) and a novel duplication (p.Glu434_Leu436dup). The detection of GOM by EM in skin biopsies was a highly reliable diagnostic method: in this cohort the congruence between NOTCH3 mutations and presence of GOM was 100%. However, due to the retrospective nature of this study, exact figure for sensitivity cannot be determined, but it would require a prospective study to exclude possible selection bias. The identification of a pathogenic NOTCH3 mutation is an indisputable evidence for CADASIL, but demonstration of GOM provides a cost-effective guide for estimating how far one should proceed with the extensive search for a new or an uncommon mutations among the presently known over 170 different NOTCH3 gene defects. The diagnostic skin biopsy should include the border zone between deep dermis and upper subcutis, where small arterial vessels of correct size are located. Detection of GOM requires technically adequate biopsies and distinction of true GOM from fallacious deposits. If GOM is not found in the first vessel or biopsy, other vessels or additional biopsies should be examined.


Asunto(s)
CADASIL/genética , Mutación , Receptores Notch/genética , Arteriolas/ultraestructura , Biopsia , CADASIL/patología , Gránulos Citoplasmáticos , Humanos , Microscopía Electrónica , Músculo Liso Vascular/ultraestructura , Receptor Notch3 , Estudios Retrospectivos , Piel/irrigación sanguínea , Piel/ultraestructura , Vénulas/ultraestructura
10.
Hum Mutat ; 29(3): 452, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18273901

RESUMEN

The most common causative diagnosis of hereditary small-vessel-disease of the brain, CADASIL, is due to highly stereotyped mutations in the NOTCH3 receptor. NOTCH3 has 33 exons but all CADASIL mutations occur within the Epidermal Growth Factor-like Repeats encoded by exons 2-24, lead to an odd number of cysteine residues and are associated with GOM deposits and abnormal NOTCH3 protein accumulation. The majority of CADASIL mutations appear to retain normal level of signaling activity, while very few mutations show reduced activity. Herein we identified a novel heterozygous missense mutation (c.4544T>C) in exon 25 of NOTCH3 in a patient with cerebral small-vessel-disease but lacking GOM deposits and NOTCH3 accumulation. The mutation should result in a p.L1515P substitution in the evolutionarily highly conserved juxtamembranous region of NOTCH3, which constitutes the heterodimerization domain. The p.L1515P mutant exhibits increased canonical NOTCH3 signaling, although in a ligand-independent fashion. Biochemical analysis suggests that the mutation renders NOTCH3 hyperactive through destabilization of the heterodimer. Therefore, our study suggests that the p.L1515P mutation falls in a novel mechanistic class of NOTCH3 mutations and that NOTCH3 activating mutations should be further considered for molecular analysis of patients with cerebral small-vessel-disease.


Asunto(s)
CADASIL/genética , Mutación Missense , Receptores Notch/genética , Sustitución de Aminoácidos , Animales , CADASIL/metabolismo , CADASIL/patología , Dimerización , Femenino , Heterocigoto , Humanos , Ratones , Persona de Mediana Edad , Células 3T3 NIH , Estructura Cuaternaria de Proteína , Receptor Notch3 , Receptores Notch/química , Receptores Notch/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección
11.
Hum Mol Genet ; 16(8): 982-92, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17331978

RESUMEN

Cerebral Autosomal Dominant Arteriopathy with Subcortical infarcts and Leukoencephalopathy (CADASIL) is the most prominent known cause of inherited stroke and vascular dementia in human adult. The disease gene, NOTCH3, encodes a transmembrane receptor primarily expressed in arterial smooth muscle cells (SMC). Pathogenic mutations lead to an odd number of cysteine residues within the NOTCH3 extracellular domain (NOTCH3(ECD)), and are associated with progressive accumulation of NOTCH3(ECD) at the SMC plasma membrane. The murine homolog, Notch3, is dispensable for viability but required post-natally for the elaboration and maintenance of arteries. How CADASIL-associated mutations impact NOTCH3 function remains a fundamental, yet unresolved issue. Particularly, whether NOTCH3(ECD) accumulation may titrate the ligand and inhibit the normal pathway is unknown. Herein, using genetic analyses in the mouse, we assessed the functional significance of an archetypal CADASIL-associated mutation (R90C), in vivo, in brain arteries. We show that transgenic mouse lines expressing either the wild-type human NOTCH3 or the mutant R90C human NOTCH3, at comparable and physiological levels, can rescue the arterial defects of Notch3-/- mice to similar degrees. In vivo assessment of NOTCH3/RBP-Jk activity provides evidence that the mutant NOTCH3 protein exhibits normal level of activity in brain arteries. Remarkably, the mutant NOTCH3 protein remains functional and does not exhibit dominant negative interfering activity, even when NOTCH3(ECD) accumulates. Collectively, these data suggest a model that invokes novel pathogenic roles for the mutant NOTCH3 protein rather than compromised NOTCH3 function as the primary determinant of the CADASIL arteriopathy.


Asunto(s)
CADASIL/genética , Receptores Notch/genética , Receptores Notch/fisiología , Envejecimiento/genética , Animales , Arginina/genética , CADASIL/patología , Arterias Cerebrales/metabolismo , Cisteína/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Enfermedades Arteriales Intracraneales/genética , Enfermedades Arteriales Intracraneales/prevención & control , Operón Lac , Ratones , Ratones Noqueados , Proteínas Mutantes/fisiología , Mutación Missense , Estructura Terciaria de Proteína/genética , Receptor Notch3 , Receptores Notch/metabolismo , Transgenes/fisiología
12.
Intern Med ; 45(16): 981-5, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16974063

RESUMEN

We report a 38-year-old Japanese woman who had cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) with a novel mutation (TGT to TAT) at nucleotide position 1241 (C388Y) in exon 7 of the Notch3 gene (NOTCH3). Immunostaining of a skin biopsy with a Notch3 monoclonal antibody is a beneficial method for the screening of CADASIL, particularly in the case of rare mutations outside the mutation hotspots in NOTCH3 as shown in this patient.


Asunto(s)
CADASIL/genética , Exones , Mutación , Receptores Notch/genética , Adulto , Pueblo Asiatico/genética , Secuencia de Bases , Biopsia , CADASIL/diagnóstico , CADASIL/metabolismo , CADASIL/patología , Femenino , Humanos , Inmunohistoquímica , Imagen por Resonancia Magnética , Microscopía Electrónica , Receptor Notch3 , Piel/metabolismo , Piel/patología
13.
Ann Neurol ; 59(2): 353-7, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16404745

RESUMEN

OBJECTIVE: Only few hereditary ischemic small vessel diseases of the brain (SVDB) have been reported so far. Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most frequent of them. Herein, we report a family affected by a SVDB distinct from CADASIL. METHODS: After the occurrence of a small deep infarct associated with white matter lesions both in a 46-year-old man and in his 52-year-old sister, clinical and neuroimaging investigations were conducted in 13 of their relatives originating from Portugal. Other investigations included (1) skin biopsy immunostaining with a Notch3 monoclonal antibody, (2) sequencing of the 23 exons encoding the epidermal growth factor-like domains of the NOTCH3 gene, and (3) a NOTCH3 locus haplotype analysis. RESULTS: Diffuse white matter hyperintensities were observed on T2-weighted magnetic resonance imaging (MRI) in six individuals. In contrast with MRI results in the father and paternal uncle of the proband who were hypertensive, white matter lesions were extensive in the mother who had no vascular risk factor. MRI data in four asymptomatic family members together with the results in the two initial cases were suggestive of an underlying hereditary small vessel disease of the brain. Skin biopsy and NOTCH3 gene mutation screening were negative. Haplotype analysis excluded the NOTCH3 locus. INTERPRETATION: These data strongly suggest that this family is affected by a novel hereditary small vessel disease of the brain and that the mutated gene is distinct from NOTCH3.


Asunto(s)
Isquemia Encefálica/genética , Receptores Notch/genética , Isquemia Encefálica/clasificación , Isquemia Encefálica/diagnóstico , Isquemia Encefálica/patología , Mapeo Encefálico , CADASIL/diagnóstico , Salud de la Familia , Femenino , Humanos , Angiografía por Resonancia Magnética/métodos , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Receptor Notch3 , Receptores Notch/metabolismo , Piel/metabolismo , Piel/patología
14.
Stroke ; 36(5): 1053-8, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15817893

RESUMEN

BACKGROUND AND PURPOSE: Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is an inherited small vessel disease causing stroke and dementia. The disease is caused by highly stereotyped mutations in NOTCH3, which is restrictively expressed in vascular smooth muscle cells (VSMCs). The mechanisms of compromised cerebral hemodynamics in CADASIL remain to be elucidated. We tested the hypothesis that mutant NOTCH3 impairs the vasomotor function of cerebral vessels. METHODS: Vasomotor function was examined in vivo in transgenic mice expressing a mutant NOTCH3 in VSMCs (TgNotch3R90C). Mice develop an age-dependent arteriopathy similar to that seen in CADASIL, without brain parenchyma lesions. Using laser-Doppler flowmetry, we assessed in awake TgNotch3R90C mice and wild-type littermates the cerebrovascular reactivity to 2 potent vasodilator stimuli (acetazolamide and hypercapnia) and cerebral blood flow (CBF) autoregulation during stepwise blood pressure elevations and reductions. Mice were studied at 18 months of age, when the CADASIL features are apparent, and at 10 months of age, before their appearance. RESULTS: Eighteen-month-old TgNotch3R90C mice showed reduced responses to hypercapnia and acetazolamide, higher cerebrovascular resistance during hypertension, and their lower limit of CBF autoregulation was shifted to higher blood pressures. Cerebrovascular responses were similarly impaired in 10-month-old TgNotch3R90C mice. CONCLUSIONS: Cerebrovascular reactivity is compromised early in TgNotch3R90C mice. The data show an impaired autoregulation and are suggestive of a decreased relaxation or increased resistance of cerebral vessels. Our findings indicate that vascular dysfunction is an early pathogenic event that may promote the subsequent development of brain ischemia in CADASIL.


Asunto(s)
CADASIL/fisiopatología , Circulación Cerebrovascular , Proteínas Proto-Oncogénicas/genética , Receptores de Superficie Celular/genética , Acetazolamida/farmacología , Animales , Presión Sanguínea , CADASIL/metabolismo , Modelos Animales de Enfermedad , Femenino , Homeostasis , Hipercapnia/fisiopatología , Flujometría por Láser-Doppler , Masculino , Ratones , Ratones Transgénicos , Músculo Liso Vascular/metabolismo , Mutación , Proteínas Proto-Oncogénicas/metabolismo , Receptor Notch3 , Receptor Notch4 , Receptores de Superficie Celular/metabolismo , Receptores Notch , Vasodilatación
15.
Stroke ; 36(1): 113-7, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15569862

RESUMEN

BACKGROUND AND PURPOSE: CADASIL is an inherited small-vessel disease responsible for lacunar strokes and cognitive impairment. The disease is caused by highly stereotyped mutations in Notch3, the expression of which is highly restricted to vascular smooth muscle cells (VSMCs). The underlying vasculopathy is characterized by degeneration of VSMCs and the accumulation of granular osmiophilic material (GOM) and Notch3 protein within the cell surface of these cells. In this study, we assessed early functional changes related to the expression of mutant Notch3 in resistance arteries. METHODS: Vasomotor function was examined in vitro in arteries from transgenic mice that express a mutant Notch3 in VSMC. Tail artery segments from transgenic and normal wild-type male mice were mounted on small-vessel arteriographs, and reactivity to mechanical (flow and pressure) forces and pharmacological stimuli were determined. Mice were studied at 10 to 11 months of age when VSMC degeneration, GOM deposits, and Notch3 accumulation were not yet present. RESULTS: Passive arterial diameter, contraction to phenylephrine, and endothelium-dependent relaxation to acetylcholine were unaffected in transgenic mice. By contrast, flow-induced dilation was significantly decreased and pressure-induced myogenic tone significantly increased in arteries from transgenic mice compared with wild-type mice. CONCLUSIONS: This is the first study to our knowledge providing evidence that mutant Notch3 impairs selectively the response of resistance arteries to flow and pressure. The data suggest an early role of vascular dysfunction in the pathogenic process of the disease.


Asunto(s)
CADASIL/fisiopatología , Mecanotransducción Celular , Proteínas Proto-Oncogénicas/genética , Receptores de Superficie Celular/genética , Vasoconstricción , Vasodilatación , Animales , Arterias/efectos de los fármacos , Arterias/fisiopatología , CADASIL/etiología , Modelos Animales de Enfermedad , Técnicas In Vitro , Masculino , Ratones , Ratones Transgénicos , Mutación , Fenilefrina/farmacología , Presión , Receptor Notch3 , Receptor Notch4 , Receptores Notch , Estrés Mecánico , Vasoconstrictores/farmacología
16.
Genes Dev ; 18(22): 2730-5, 2004 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-15545631

RESUMEN

Formation of a fully functional artery proceeds through a multistep process. Here we show that Notch3 is required to generate functional arteries in mice by regulating arterial differentiation and maturation of vascular smooth muscle cells (vSMC). In adult Notch3-/- mice distal arteries exhibit structural defects and arterial myogenic responses are defective. The postnatal maturation stage of vSMC is deficient in Notch3-/- mice. We further show that Notch3 is required for arterial specification of vSMC but not of endothelial cells. Our data reveal Notch3 to be the first cell-autonomous regulator of arterial differentiation and maturation of vSMC.


Asunto(s)
Diferenciación Celular , Células Endoteliales/citología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Proteínas Proto-Oncogénicas/fisiología , Receptores de Superficie Celular/fisiología , Actinas/metabolismo , Animales , Velocidad del Flujo Sanguíneo , Presión Sanguínea , Células Cultivadas , Desmina/metabolismo , Células Endoteliales/metabolismo , Homocigoto , Humanos , Hibridación in Situ , Operón Lac/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/fisiología , Proteínas Musculares/genética , Proteínas Musculares/fisiología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Proto-Oncogénicas/genética , Receptor Notch3 , Receptor Notch4 , Receptores de Superficie Celular/genética , Receptores Notch , Porcinos
17.
Ann Neurol ; 55(2): 213-20, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14755725

RESUMEN

Cerebral Cavernous Malformations (CCM/OMIM 604214) are vascular malformations causing seizures and cerebral hemorrhages. They occur as a sporadic and autosomal dominant condition, the latter being characterized by the presence of multiple CCM lesions. Stereotyped truncating mutations of KRIT1, the sole CCM gene identified so far, have been identified in CCM1 linked families but the clinical features associated with KRIT1 mutations have not yet been assessed in a large series of patients. We conducted a detailed clinical, neuroradiological and molecular analysis of 64 consecutively recruited CCM families segregating a KRIT1 mutation. Those families included 202 KRIT1 mutation carriers. Among the 202 KRIT1 mutation carriers, 126 individuals were symptomatic and 76 symptom-free. Mean age at clinical onset was 29.7 years (range, 2-72); initial clinical manifestations were seizures in 55% of the cases and cerebral hemorrhages in 32%. Average number of lesions on T2 weighted MRI was 4.9 (+/-7.2) and on gradient echo sequences 19.8 (+/-33.2). Twenty-six mutation carriers harbored only one lesion on T2-weighted MRI, including 4 mutation carriers, aged from 18 to 55 yr-old, who presented only one CCM lesion both on T2-weighted and on highly sensitive gradient echo MRI sequences. Five symptom free mutation carriers, aged from 27 to 48 yr-old, did not have any detectable lesion both on T2WI and gradient echo MRI sequences. Within KRIT1/CCM1 families, both clinical and radiological penetrance are incomplete and age dependent. Importantly for genetic counseling, nearly half of the KRIT1 mutation carriers aged 50 or more are symptom-free. The presence of only one lesion, even when using gradient echo MRI sequences, can be observed in some patients with an hereditary form of the disease. Incomplete neuroradiological penetrance precludes the use of cerebral MRI to firmly establish a non carrier status, even at an adult age and even when using highly sensitive gradient echo MRI. Altogether these data suggest that the hereditary nature of the disorder may be overlooked in some mutation carriers presenting as sporadic cases with a unique lesion.


Asunto(s)
Encéfalo/patología , Hemangioma Cavernoso del Sistema Nervioso Central/genética , Hemangioma Cavernoso del Sistema Nervioso Central/patología , Proteínas Asociadas a Microtúbulos/genética , Penetrancia , Proteínas Proto-Oncogénicas/genética , Adolescente , Adulto , Anciano , Hemorragia Cerebral/etiología , Niño , Preescolar , Femenino , Hemangioma Cavernoso del Sistema Nervioso Central/complicaciones , Heterocigoto , Humanos , Lactante , Proteína KRIT1 , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Mutación , Linaje , Polimorfismo Conformacional Retorcido-Simple , Convulsiones/etiología , Factores Sexuales
18.
Am J Hum Genet ; 74(2): 338-47, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14714274

RESUMEN

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is an inherited vascular dementia characterized by the degeneration of smooth-muscle cells in small cerebral arteries. CADASIL is caused by mutations in NOTCH3, one of the four mammalian homologs to the Drosophila melanogaster NOTCH gene. Disease-associated mutations are distributed throughout the 34 epidermal growth factor-like repeats (EGFRs) that compose the extracellular domain of the Notch3 receptor and result in a loss or a gain of a cysteine residue in one of these EGFRs. In human adults, Notch3 expression is highly restricted to vascular smooth-muscle cells. In patients with CADASIL, there is an abnormal accumulation of Notch3 in the vessel. Molecular pathways linking NOTCH3 mutations to degeneration of vascular smooth-muscle cells are as yet poorly understood. In this study, we investigated the effect of CADASIL mutations on Notch3 activity. We studied five naturally occurring mutations: R90C and C212S, located in the previously identified mutational hotspot EGFR2-5; C428S, shown in this study to be located in the ligand-binding domain EGFR10-11; and C542Y and R1006C, located in EGFR13 and EGFR26, respectively. All five mutant proteins were correctly processed. The C428S and C542Y mutant receptors exhibited a significant reduction in Jagged1-induced transcriptional activity of a RBP/JK responsive luciferase reporter, relative to wild-type Notch3. Impaired signaling activity of these two mutants arose through different mechanisms; the C428S mutant lost its Jagged1-binding ability, whereas C542Y retained it but exhibited an impaired presentation to the cell surface. In contrast, the R90C, C212S, and R1006C mutants retained the ability to bind Jagged1 and were associated with apparently normal levels of signaling activity. We conclude that mutations in Notch3 differently affect Jagged1 binding and Notch3 signaling via the RBP/JK pathway.


Asunto(s)
Demencia por Múltiples Infartos/genética , Mutación , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Superficie Celular , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas de Unión al Calcio , Proteínas de Drosophila , Humanos , Péptidos y Proteínas de Señalización Intercelular , Proteína Jagged-1 , Proteínas de la Membrana , Unión Proteica , Receptor Notch3 , Receptores Notch , Proteínas Serrate-Jagged
19.
Genesis ; 37(3): 139-43, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14595837

RESUMEN

The Notch signaling pathway is an evolutionarily conserved signaling mechanism and mutations in its components disrupt cell fate specification and embryonic development in many organisms. To analyze the in vivo role of the Notch3 gene in mice, we created a deletion allele by gene targeting. Embryos homozygous for this mutation developed normally and homozygous mutant adults were viable and fertile. We also examined whether we could detect genetic interactions during early embryogenesis between the Notch3 mutation and a targeted mutation of the Notch1 gene. Double homozygous mutant embryos exhibited defects normally observed in Notch1-deficient embryos, but we detected no obvious synergistic effects in the double mutants. These data demonstrate that the Notch3 gene is not essential for embryonic development or fertility in mice, and does not have a redundant function with the Notch1 gene during early embryogenesis.


Asunto(s)
Desarrollo Embrionario y Fetal/genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/farmacología , Receptores de Superficie Celular/genética , Factores de Transcripción , Animales , Femenino , Fertilidad/genética , Vectores Genéticos , Masculino , Ratones/embriología , Mutación , Receptor Notch1 , Receptor Notch3 , Receptor Notch4 , Receptores de Superficie Celular/fisiología , Receptores Notch , Transducción de Señal
20.
Am J Pathol ; 162(1): 329-42, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12507916

RESUMEN

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is an increasingly recognized adult-onset autosomal dominant vascular dementia, caused by highly stereotyped mutations in the Notch3 receptor. CADASIL is a widespread angiopathy characterized by a degeneration of vascular smooth muscle cells (VSMCs) and the abnormal accumulation of electron-dense granular material called GOM and Notch3 protein, because of an impaired clearance. Evidence that VSMCs are the primary target of the pathogenic process is supported by the restricted expression of Notch3 in these cells but mechanisms of their degeneration remain essentially unknown. We generated transgenic mice in which the SM22alpha promoter drove, in VSMCs, the expression of a full-length human Notch3 carrying the Arg90Cys mutation, a CADASIL archetypal mutation. Transgenic mice showed no evidence of prominent brain parenchyma damage but demonstrated the two hallmarks of the CADASIL angiopathy, GOM deposits and Notch3 accumulation, within both the cerebral and peripheral arteries. Of interest, arteries of the tail were more severely affected with prominent signs of VSMC degeneration. Time-course analysis of vessel changes revealed that disruption of normal VSMC anchorage to adjacent extracellular matrix and cells, VSMC cytoskeleton changes as well as starting signs of VSMC degeneration, which were detected around 10 months of age, preceded Notch3 and GOM accumulation appearance, which were observed only by 14 to 16 months of age. In conclusion, we have generated transgenic mice that recapitulate the characteristic vascular lesions observed in CADASIL. Our results indicate that Notch3 or GOM accumulation are unlikely to be the prerequisites for the induction of VSMC degeneration and suggest that degeneration of VSMCs may rather be triggered by the disruption of their normal anchorage, based on the important role of adhesion for cell survival.


Asunto(s)
Demencia por Múltiples Infartos/genética , Demencia por Múltiples Infartos/patología , Modelos Animales de Enfermedad , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Receptores de Superficie Celular , Factores de Edad , Sustitución de Aminoácidos , Animales , Arterias/patología , Vasos Sanguíneos/patología , Vasos Sanguíneos/ultraestructura , Encéfalo/irrigación sanguínea , Encéfalo/patología , Progresión de la Enfermedad , Humanos , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Proteínas Musculares/genética , Músculo Liso Vascular/patología , Mutación , Regiones Promotoras Genéticas , Receptor Notch3 , Receptor Notch4 , Receptores Notch , Cola (estructura animal)/irrigación sanguínea , Cola (estructura animal)/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA