Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Cell Dev Biol ; 11: 1252547, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37691820

RESUMEN

Rare DRAM2 coding variants cause retinal dystrophy with early macular involvement via unknown mechanisms. We found that DRAM2 is ubiquitously expressed in the human eye and expression changes were observed in eyes with more common maculopathy such as Age-related Macular Degeneration (AMD). To gain insights into pathogenicity of DRAM2-related retinopathy, we used a combination of in vitro and in vivo models. We found that DRAM2 loss in human pluripotent stem cell (hPSC)-derived retinal organoids caused the presence of additional mesenchymal cells. Interestingly, Dram2 loss in mice also caused increased proliferation of cells from the choroid in vitro and exacerbated choroidal neovascular lesions in vivo. Furthermore, we observed that DRAM2 loss in human retinal pigment epithelial (RPE) cells resulted in increased susceptibility to stress-induced cell death in vitro and that Dram2 loss in mice caused age-related photoreceptor degeneration. This highlights the complexity of DRAM2 function, as its loss in choroidal cells provided a proliferative advantage, whereas its loss in post-mitotic cells, such as photoreceptor and RPE cells, increased degeneration susceptibility. Different models such as human pluripotent stem cell-derived systems and mice can be leveraged to study and model human retinal dystrophies; however, cell type and species-specific expression must be taken into account when selecting relevant systems.

2.
PLoS One ; 18(4): e0280484, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37079518

RESUMEN

BACKGROUND: The basis of Age-related macular degeneration (AMD) genetic risk has been well documented; however, few studies have looked at genetic biomarkers of disease progression or treatment response within advanced AMD patients. Here we report the first genome-wide analysis of genetic determinants of low-luminance vision deficit (LLD), which is seen as predictive of visual acuity loss and anti-VEGF treatment response in neovascular AMD patients. METHODS: AMD patients were separated into small- and large-LLD groups for comparison and whole genome sequencing was performed. Genetic determinants of LLD were assessed by common and rare variant genetic analysis. Follow-up functional analysis of rare coding variants identified by the burden test was then performed in vitro. RESULTS: We identified four coding variants in the CIDEC gene. These rare variants were only present in patients with a small LLD, which has been previously shown to indicate better prognosis and better anti-VEGF treatment response. Our in vitro functional characterization of these CIDEC alleles revealed that all decrease the binding affinity between CIDEC and the lipid droplet fusion effectors PLIN1, RAB8A and AS160. The rare CIDEC alleles all cause a hypomorphic defect in lipid droplet fusion and enlargement, resulting in a decreased fat storage capability in adipocytes. CONCLUSIONS: As we did not detect CIDEC expression in the ocular tissue affected by AMD, our results suggest that the CIDEC variants do not play a direct role in the eye and influence low-luminance vision deficit via an indirect and systemic effect related to fat storage capacity.


Asunto(s)
Baja Visión , Degeneración Macular Húmeda , Humanos , Inhibidores de la Angiogénesis , Gotas Lipídicas/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Agudeza Visual/genética , Degeneración Macular Húmeda/metabolismo
4.
Dis Model Mech ; 14(4)2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-34424299

RESUMEN

Collagen type IV alpha 1 and alpha 2 (COL4A1 and COL4A2) are major components of almost all basement membranes. COL4A1 and COL4A2 mutations cause a multisystem disorder that can affect any organ but typically involves the cerebral vasculature, eyes, kidneys and skeletal muscles. In recent years, patient advocacy and family support groups have united under the name of Gould syndrome. The manifestations of Gould syndrome are highly variable, and animal studies suggest that allelic heterogeneity and genetic context contribute to the clinical variability. We previously characterized a mouse model of Gould syndrome caused by a Col4a1 mutation in which the severities of ocular anterior segment dysgenesis (ASD), myopathy and intracerebral hemorrhage (ICH) were dependent on genetic background. Here, we performed a genetic modifier screen to provide insight into the mechanisms contributing to Gould syndrome pathogenesis and identified a single locus [modifier of Gould syndrome 1 (MoGS1)] on Chromosome 1 that suppressed ASD. A separate screen showed that the same locus ameliorated myopathy. Interestingly, MoGS1 had no effect on ICH, suggesting that this phenotype could be mechanistically distinct. We refined the MoGS1 locus to a 4.3 Mb interval containing 18 protein-coding genes, including Fn1, which encodes the extracellular matrix component fibronectin 1. Molecular analysis showed that the MoGS1 locus increased Fn1 expression, raising the possibility that suppression is achieved through a compensatory extracellular mechanism. Furthermore, we found evidence of increased integrin-linked kinase levels and focal adhesion kinase phosphorylation in Col4a1 mutant mice that is partially restored by the MoGS1 locus, implicating the involvement of integrin signaling. Taken together, our results suggest that tissue-specific mechanistic heterogeneity contributes to the variable expressivity of Gould syndrome and that perturbations in integrin signaling may play a role in ocular and muscular manifestations.


Asunto(s)
Anomalías Múltiples/genética , Colágeno Tipo IV/genética , Fibronectinas/genética , Genes Modificadores , Animales , Hemorragia Cerebral/complicaciones , Mapeo Cromosómico , Cromosomas de los Mamíferos/genética , Anomalías del Ojo/complicaciones , Anomalías del Ojo/genética , Fibronectinas/metabolismo , Genes Supresores , Sitios Genéticos , Integrinas/metabolismo , Ratones Mutantes , Enfermedades Musculares/genética , Porencefalia/complicaciones , Transducción de Señal , Síndrome
5.
Cell Rep ; 30(4): 1246-1259.e6, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31995762

RESUMEN

Age-related macular degeneration (AMD) is a leading cause of vision loss. To better understand disease pathogenesis and identify causal genes in GWAS loci for AMD risk, we present a comprehensive database of human retina and retinal pigment epithelium (RPE). Our database comprises macular and non-macular RNA sequencing (RNA-seq) profiles from 129 donors, a genome-wide expression quantitative trait loci (eQTL) dataset that includes macula-specific retina and RPE/choroid, and single-nucleus RNA-seq (NucSeq) from human retina and RPE with subtype resolution from more than 100,000 cells. Using NucSeq, we find enriched expression of AMD candidate genes in RPE cells. We identify 15 putative causal genes for AMD on the basis of co-localization of genetic association signals for AMD risk and eye eQTL, including the genes TSPAN10 and TRPM1. These results demonstrate the value of our human eye database for elucidating genetic pathways and potential therapeutic targets for ocular diseases.


Asunto(s)
Susceptibilidad a Enfermedades/metabolismo , Regulación de la Expresión Génica/genética , Degeneración Macular/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Anciano , Anciano de 80 o más Años , Alelos , Coroides/metabolismo , Bases de Datos Genéticas , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Degeneración Macular/genética , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , RNA-Seq , Factores de Riesgo , Análisis de la Célula Individual , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/metabolismo , Tetraspaninas/genética , Tetraspaninas/metabolismo , Transcriptoma/genética
6.
Matrix Biol ; 57-58: 29-44, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27794444

RESUMEN

COL4A1 and COL4A2 are extracellular matrix proteins that form heterotrimers and are present in nearly all basement membranes in every organ. In the past decade, COL4A1 and COL4A2 mutations have been identified to cause a multi-system disorder for which penetrance and severity of constituent phenotypes can greatly vary. Here, we compare the outcomes of more than 100 mutations identified in patients and data from a murine allelic series to explore the presence of genotype-phenotype correlations - many of which are shared among other types of collagen. We find that there is a frequency bias for COL4A1 over COL4A2 mutations and that glycine (Gly) substitutions within the triple helical domain are the most common class of mutations. Glycine is most often replaced by a charged amino acid, however the position of the mutation, and not the properties of the substituting amino acid, appears to have a greater influence on disease severity. Moreover, the impact of position is not straightforward. Observations from a murine allelic series suggest that mutations in the NC1 domain may result in relatively mild phenotypes via a 'quantitative' mechanism similar to other types of collagens, however, this effect was not apparent in human reports. Importantly, other position-dependent effects had differential impacts depending on the phenotype of interest. For example, the severity of cerebrovascular disease correlated with an amino-to-carboxy severity gradient for triple-helical glycine substitutions whereas the penetrance and severity of myopathy and nephropathy appear to involve a functional sub-domain(s). Greater understanding of genotype-phenotype correlations and the interaction of consequences of different mutations will be important for patient prognosis and care and for developing mechanism-based therapeutics to treat individual components of this emerging syndrome.


Asunto(s)
Membrana Basal/metabolismo , Trastornos Cerebrovasculares/genética , Colágeno Tipo IV/genética , Estudios de Asociación Genética , Enfermedades Renales/genética , Enfermedades Musculares/genética , Sustitución de Aminoácidos , Animales , Membrana Basal/química , Membrana Basal/patología , Trastornos Cerebrovasculares/metabolismo , Trastornos Cerebrovasculares/patología , Colágeno Tipo IV/metabolismo , Regulación de la Expresión Génica , Glicina/química , Glicina/metabolismo , Humanos , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Ratones , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Mutación , Dominios Proteicos , Transducción de Señal
8.
J Clin Invest ; 126(10): 3758-3771, 2016 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-27571405

RESUMEN

Studies of the genetic factors associated with human autoimmune disease suggest a multigenic origin of susceptibility; however, how these factors interact and through which tolerance pathways they operate generally remain to be defined. One key checkpoint occurs through the activity of the autoimmune regulator AIRE, which promotes central T cell tolerance. Recent reports have described a variety of dominant-negative AIRE mutations that likely contribute to human autoimmunity to a greater extent than previously thought. In families with these mutations, the penetrance of autoimmunity is incomplete, suggesting that other checkpoints play a role in preventing autoimmunity. Here, we tested whether a defect in LYN, an inhibitory protein tyrosine kinase that is implicated in systemic autoimmunity, could combine with an Aire mutation to provoke organ-specific autoimmunity. Indeed, mice with a dominant-negative allele of Aire and deficiency in LYN spontaneously developed organ-specific autoimmunity in the eye. We further determined that a small pool of retinal protein-specific T cells escaped thymic deletion as a result of the hypomorphic Aire function and that these cells also escaped peripheral tolerance in the presence of LYN-deficient dendritic cells, leading to highly destructive autoimmune attack. These findings demonstrate how 2 distinct tolerance pathways can synergize to unleash autoimmunity and have implications for the genetic susceptibility of autoimmune disease.


Asunto(s)
Autoinmunidad , Factores de Transcripción/fisiología , Familia-src Quinasas/fisiología , Animales , Presentación de Antígeno , Autoanticuerpos/metabolismo , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Proteínas del Ojo/inmunología , Microbioma Gastrointestinal/inmunología , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos , Proteínas de Unión al Retinol/inmunología , Uveítis Posterior/genética , Uveítis Posterior/inmunología , Proteína AIRE
9.
BMC Biol ; 14: 59, 2016 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-27412481

RESUMEN

BACKGROUND: Type IV collagen is the main component of the basement membrane that gives strength to the blood-gas barrier (BGB). In mammals, the formation of a mature BGB occurs primarily after birth during alveologenesis and requires the formation of septa from the walls of the saccule. In contrast, in avians, the formation of the BGB occurs rapidly and prior to hatching. Mutation in basement membrane components results in an abnormal alveolar phenotype; however, the specific role of type IV collagen in regulating alveologenesis remains unknown. RESULTS: We have performed a microarray expression analysis in late chick lung development and found that COL4A1 and COL4A2 were among the most significantly upregulated genes during the formation of the avian BGB. Using mouse models, we discovered that mutations in murine Col4a1 and Col4a2 genes affected the balance between lung epithelial progenitors and differentiated cells. Mutations in Col4a1 derived from the vascular component were sufficient to cause defects in vascular development and the BGB. We also show that Col4a1 and Col4a2 mutants displayed disrupted myofibroblast proliferation, differentiation and migration. Lastly, we revealed that addition of type IV collagen protein induced myofibroblast proliferation and migration in monolayer culture and increased the formation of mesenchymal-epithelial septal-like structures in co-culture. CONCLUSIONS: Our study showed that type IV collagen and, therefore the basement membrane, play fundamental roles in coordinating alveolar morphogenesis. In addition to its role in the formation of epithelium and vasculature, type IV collagen appears to be key for alveolar myofibroblast development by inducing their proliferation, differentiation and migration throughout the developing septum.


Asunto(s)
Colágeno Tipo IV/metabolismo , Células Endoteliales/citología , Células Epiteliales/citología , Morfogénesis , Fragmentos de Péptidos/metabolismo , Células A549 , Animales , Membrana Basal/metabolismo , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Embrión de Pollo , Técnicas de Cocultivo , Colágeno Tipo IV/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Pulmón/citología , Ratones , Ratones Noqueados , Análisis por Micromatrices , Mutación , Miofibroblastos/citología , Fragmentos de Péptidos/genética , Regulación hacia Arriba
10.
Circulation ; 131(18): 1555-65, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25753534

RESUMEN

BACKGROUND: Collagen type IV alpha1 (COL4A1) and alpha2 (COL4A2) form heterotrimers critical for vascular basement membrane stability and function. Patients with COL4A1 or COL4A2 mutations suffer from diverse cerebrovascular diseases, including cerebral microbleeds, porencephaly, and fatal intracerebral hemorrhage (ICH). However, the pathogenic mechanisms remain unknown, and there is a lack of effective treatment. METHODS AND RESULTS: Using Col4a1 and Col4a2 mutant mouse models, we investigated the genetic complexity and cellular mechanisms underlying the disease. We found that Col4a1 mutations cause abnormal vascular development, which triggers small-vessel disease, recurrent hemorrhagic strokes, and age-related macroangiopathy. We showed that allelic heterogeneity, genetic context, and environmental factors such as intense exercise or anticoagulant medication modulated disease severity and contributed to phenotypic heterogeneity. We found that intracellular accumulation of mutant collagen in vascular endothelial cells and pericytes was a key triggering factor of ICH. Finally, we showed that treatment of mutant mice with a US Food and Drug Administration-approved chemical chaperone resulted in a decreased collagen intracellular accumulation and a significant reduction in ICH severity. CONCLUSIONS: Our data are the first to show therapeutic prevention in vivo of ICH resulting from Col4a1 mutation and imply that a mechanism-based therapy promoting protein folding might also prevent ICH in patients with COL4A1 and COL4A2 mutations.


Asunto(s)
Hemorragia Cerebral/prevención & control , Colágeno Tipo IV/genética , Heterogeneidad Genética , Fragmentos de Péptidos/genética , Animales , Vasos Sanguíneos/anomalías , Vasos Sanguíneos/embriología , Barrera Hematoencefálica , Encéfalo/irrigación sanguínea , Encéfalo/embriología , Hemorragia Cerebral/genética , Colágeno/metabolismo , Colágeno Tipo IV/deficiencia , Colágeno Tipo IV/fisiología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Femenino , Interacción Gen-Ambiente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Neovascularización Fisiológica/genética , Fragmentos de Péptidos/deficiencia , Fragmentos de Péptidos/fisiología , Pericitos/metabolismo , Fenotipo , Condicionamiento Físico Animal , Porencefalia/genética , Vasos Retinianos/embriología
11.
Am J Pathol ; 184(6): 1695-705, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24736166

RESUMEN

Development of novel strategies to treat noninfectious posterior uveitis is an ongoing challenge, in part because of limited availability of animal models that mimic the naturally occurring disease in humans. Mice deficient in the autoimmune regulatory gene Aire develop a spontaneous T-cell and macrophage-mediated autoimmune uveitis that closely recapitulates human endogenous uveitis and thus provide a useful model for mechanistic and therapeutic investigations. Lymphocytic and mononuclear infiltration of the retina in Aire knockout (KO) mice triggers the onset of uveitis from initial retinal inflammation to eventual destruction of the neuroretina with loss of photoreceptors. The C-C chemokine receptor type 2 protein (CCR2) functions in directing monocyte and macrophage migration to inflamed tissues via interaction with monocyte chemotactic proteins. Using the Aire KO mouse model, we demonstrated an essential role for CCR2 in the pathogenesis of autoimmune-mediated uveitis. Loss of functional CCR2 effectively reduced immune cell infiltration and rescued the retina from destruction. CCR2-dependent migration of bone marrow-derived cells provided the driving force for retinal inflammation, with CCR2-expressing mononuclear cells contributing to retinal damage via recruitment of CD4(+) T cells. These studies identify the CCR2 pathway as a promising therapeutic target that may prove an effective approach to treat uveitis associated with autoimmunity.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Movimiento Celular/inmunología , Receptores CCR2/inmunología , Retina/inmunología , Uveítis/inmunología , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Linfocitos T CD4-Positivos/patología , Movimiento Celular/genética , Humanos , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Monocitos/inmunología , Monocitos/patología , Receptores CCR2/genética , Retina/patología , Uveítis/genética , Uveítis/patología
12.
J Cell Biol ; 204(6): 931-45, 2014 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-24637324

RESUMEN

The promyelocytic leukemia (PML) protein organizes PML nuclear bodies (NBs), which are stress-responsive domains where many partner proteins accumulate. Here, we clarify the basis for NB formation and identify stress-induced partner sumoylation as the primary NB function. NB nucleation does not rely primarily on intermolecular interactions between the PML SUMO-interacting motif (SIM) and SUMO, but instead results from oxidation-mediated PML multimerization. Oxidized PML spherical meshes recruit UBC9, which enhances PML sumoylation, allow partner recruitment through SIM interactions, and ultimately enhance partner sumoylation. Intermolecular SUMO-SIM interactions then enforce partner sequestration within the NB inner core. Accordingly, oxidative stress enhances NB formation and global sumoylation in vivo. Some NB-associated sumoylated partners also become polyubiquitinated by RNF4, precipitating their proteasomal degradation. As several partners are protein-modifying enzymes, NBs could act as sensors that facilitate and confer oxidative stress sensitivity not only to sumoylation but also to other post-translational modifications, thereby explaining alterations of stress response upon PML or NB loss.


Asunto(s)
Proteínas Nucleares/metabolismo , Estrés Oxidativo , Sumoilación , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Células CHO , Células COS , Núcleo Celular/metabolismo , Senescencia Celular , Chlorocebus aethiops , Cricetinae , Cricetulus , Células HeLa , Humanos , Ratones , Proteína de la Leucemia Promielocítica , Transporte de Proteínas , Especies Reactivas de Oxígeno/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas
13.
Hum Mol Genet ; 23(7): 1709-22, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24203695

RESUMEN

Collagen type IV alpha 1 and 2 (COL4A1 and COL4A2) are present in nearly all basement membranes. COL4A1 and COL4A2 mutations are pleiotropic, affecting multiple organ systems to differing degrees, and both genetic-context and environmental factors influence this variable expressivity. Here, we report important phenotypic and molecular differences in an allelic series of Col4a1 and Col4a2 mutant mice that are on a uniform genetic background. We evaluated three organs commonly affected by COL4A1 and COL4A2 mutations and discovered allelic heterogeneity in the penetrance and severity of ocular dysgenesis, myopathy and brain malformations. Similarly, we show allelic heterogeneity in COL4A1 and COL4A2 biosynthesis. While most mutations that we examined caused increased intracellular and decreased extracellular COL4A1 and COL4A2, we identified three mutations with distinct biosynthetic signatures. Reduced temperature or presence of 4-phenylbutyrate ameliorated biosynthetic defects in primary cell lines derived from mutant mice. Together, our data demonstrate the effects and clinical implications of allelic heterogeneity in Col4a1- and Col4a2-related diseases. Understanding allelic differences will be valuable for increasing prognostic accuracy and for the development of therapeutic interventions that consider the nature of the molecular cause in patients with COL4A1 and COL4A2 mutations.


Asunto(s)
Encéfalo/anomalías , Colágeno Tipo IV/genética , Anomalías del Ojo/genética , Enfermedades Musculares/genética , Enfermedades del Nervio Óptico/congénito , Alelos , Animales , Antineoplásicos/farmacología , Membrana Basal/metabolismo , Células Cultivadas , Estrés del Retículo Endoplásmico/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Enfermedades del Nervio Óptico/genética , Penetrancia , Fenilbutiratos/farmacología , Pliegue de Proteína , Temperatura
14.
Sci Rep ; 3: 1494, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23511048

RESUMEN

Mechanical forces including gravity affect endothelial cell (ECs) function, and have been implicated in vascular disease as well as physiologic changes associated with low gravity environments. The goal of this study was to investigate the impact of gravitational mechanical unloading on ECs phenotype as determined by patterns of gene expression. Human umbilical vascular endothelial cells were exposed to 1-gravity environment or mechanical unloading (MU) for 24 hours, with or without periods of mechanical loading (ML). MU led to a significant decrease in gene expression of several adhesion molecules and pro-inflammatory cytokines. On the contrary, eNOS, Caveolin-1 and -2 expression were significantly increased with MU. There was a decrease in the length and width of the cells with MU. Addition of ML during the MU period was sufficient to reverse the changes triggered by MU. Our results suggest that gravitational loading could dramatically affect vascular endothelial cell function.


Asunto(s)
Caveolinas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Gravitación , Suspensión Trasera , Células Endoteliales de la Vena Umbilical Humana/patología , Inflamación/patología , Estrés Mecánico , Animales , Aorta/patología , Fenómenos Biomecánicos , Moléculas de Adhesión Celular/genética , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Ratones , Modelos Animales , Modelos Biológicos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Nitritos/metabolismo
15.
Ann Neurol ; 71(4): 470-7, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22522439

RESUMEN

OBJECTIVE: Mutations in the type IV collagen alpha 1 gene (COL4A1) cause dominantly inherited cerebrovascular disease. We seek to determine the extent to which COL4A1 mutations contribute to sporadic, nonfamilial, intracerebral hemorrhages (ICHs). METHODS: We sequenced COL4A1 in 96 patients with sporadic ICH. The presence of putative mutations was tested in 145 ICH-free controls. The effects of rare coding variants on COL4A1 biosynthesis were compared to previously validated mutations that cause porencephaly, small vessel disease, and hereditary angiopathy, nephropathy, aneurysms, and cramps (HANAC) syndrome. RESULTS: We identified 2 rare nonsynonymous variants in ICH patients that were not detected in controls, 2 rare nonsynonymous variants in controls that were not detected in patients, and 2 common nonsynonymous variants that were detected in patients and controls. No variant found in controls affected COL4A1 biosynthesis. Both variants (COL4A1(P352L) and COL4A1(R538G)) found only in patients changed conserved amino acids and impaired COL4A1 secretion much like mutations that cause familial cerebrovascular disease. INTERPRETATION: This is the first assessment of the broader role for COL4A1 mutations in the etiology of ICH beyond a contribution to rare and severe familial cases and the first functional evaluation of the biosynthetic consequences of an allelic series of COL4A1 mutations that cause cerebrovascular disease. We identified 2 putative mutations in 96 patients with sporadic ICH and showed that these and other previously validated mutations inhibit secretion of COL4A1. Our data support the hypothesis that increased intracellular accumulation of COL4A1, decreased extracellular COL4A1, or both, contribute to sporadic cerebrovascular disease and ICH.


Asunto(s)
Hemorragia Cerebral/genética , Colágeno Tipo IV/genética , Predisposición Genética a la Enfermedad/genética , Mutación , Anciano , Secuencia de Aminoácidos , Western Blotting , Trastornos Cerebrovasculares/genética , Análisis Mutacional de ADN , Femenino , Humanos , Masculino , Datos de Secuencia Molecular
16.
Am J Hum Genet ; 90(1): 91-101, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22209247

RESUMEN

Collagen, type IV, alpha 1 (COL4A1) and alpha 2 (COL4A2) form heterotrimers and are abundant components of basement membranes, including those of the cerebral vasculature. COL4A1 mutations are an increasingly recognized cause of multisystem disorders, including highly penetrant cerebrovascular disease and intracerebral hemorrhage (ICH). Because COL4A1 and COL4A2 are structurally and functionally associated, we hypothesized that variants in COL4A2 would also cause ICH. We sequence COL4A2 in 96 patients with ICH and identify three rare, nonsynonymous coding variants in four patients that are not present in a cohort of 144 ICH-free individuals. All three variants change evolutionarily conserved amino acids. Using a cellular assay, we show that these putative mutations cause intracellular accumulation of COL4A1 and COL4A2 at the expense of their secretion, which supports their pathogenecity. Furthermore, we show that Col4a2 mutant mice also have completely penetrant ICH and that mutations in mouse and human lead to retention of COL4A1 and COL4A2 within the endoplasmic reticulum (ER). Importantly, two of the three putative mutations found in patients trigger ER stress and activate the unfolded protein response. The identification of putative COL4A2 mutations that might contribute to ICH in human patients provides insight into the pathogenic mechanisms of this disease. Our data suggest that COL4A2 mutations impair COL4A1 and COL4A2 secretion and can also result in cytotoxicity. Finally, our findings suggest that, collectively, mutations in COL4A1 and COL4A2 contribute to sporadic cases of ICH.


Asunto(s)
Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Hemorragias Intracraneales/genética , Mutación , Accidente Cerebrovascular/genética , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Estrés del Retículo Endoplásmico/genética , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Datos de Secuencia Molecular , Respuesta de Proteína Desplegada
17.
Cancer Cell ; 20(5): 661-73, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22094259

RESUMEN

Using a mouse model recapitulating the main features of human chronic myelogenous leukemia (CML), we uncover the hierarchy of leukemic stem and progenitor cells contributing to disease pathogenesis. We refine the characterization of CML leukemic stem cells (LSCs) to the most immature long-term hematopoietic stem cells (LT-HSCs) and identify some important molecular deregulations underlying their aberrant behavior. We find that CML multipotent progenitors (MPPs) exhibit an aberrant B-lymphoid potential but are redirected toward the myeloid lineage by the action of the proinflammatory cytokine IL-6. We show that BCR/ABL activity controls Il-6 expression thereby establishing a paracrine feedback loop that sustains CML development. These results describe how proinflammatory tumor environment affects leukemic progenitor cell fate and contributes to CML pathogenesis.


Asunto(s)
Interleucina-6/fisiología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Multipotentes/patología , Animales , Retroalimentación Fisiológica , Proteínas de Fusión bcr-abl/metabolismo , Proteínas de Fusión bcr-abl/fisiología , Interleucina-6/genética , Interleucina-6/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Ratones , Células Precursoras de Linfocitos B/metabolismo , Células Precursoras de Linfocitos B/patología
18.
Cancer Cell ; 18(1): 88-98, 2010 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-20609355

RESUMEN

As(2)O(3) cures acute promyelocytic leukemia (APL) by initiating PML/RARA oncoprotein degradation, through sumoylation of its PML moiety. However, how As(2)O(3) initiates PML sumoylation has remained largely unexplained. As(2)O(3) binds vicinal cysteines and increases reactive oxygen species (ROS) production. We demonstrate that upon As(2)O(3) exposure, PML undergoes ROS-initiated intermolecular disulfide formation and binds arsenic directly. Disulfide-linked PML or PML/RARA multimers form nuclear matrix-associated nuclear bodies (NBs), become sumoylated and are degraded. Hematopoietic progenitors transformed by an As(2)O(3)-binding PML/RARA mutant exhibit defective As(2)O(3) response. Conversely, nonarsenical oxidants elicit PML/RARA multimerization, NB-association, degradation, and leukemia response in vivo, but do not affect PLZF/RARA-driven APLs. Thus, PML oxidation regulates NB-biogenesis, while oxidation-enforced PML/RARA multimerization and direct arsenic-binding cooperate to enforce APL's exquisite As(2)O(3) sensitivity.


Asunto(s)
Antineoplásicos/farmacología , Arsenicales/farmacología , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/metabolismo , Óxidos/farmacología , Animales , Trióxido de Arsénico , Western Blotting , Células CHO , Células COS , Chlorocebus aethiops , Cricetinae , Cricetulus , Disulfuros/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Cuerpos de Inclusión Intranucleares/metabolismo , Leucemia Promielocítica Aguda/patología , Ratones , Ratones Noqueados , Mutación/genética , Proteínas Nucleares/fisiología , Proteínas de Fusión Oncogénica/genética , Proteína de la Leucemia Promielocítica , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Procesamiento Proteico-Postraduccional , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Factores de Transcripción/fisiología , Proteínas Supresoras de Tumor/fisiología
19.
Science ; 328(5975): 240-3, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20378816

RESUMEN

Arsenic, an ancient drug used in traditional Chinese medicine, has attracted worldwide interest because it shows substantial anticancer activity in patients with acute promyelocytic leukemia (APL). Arsenic trioxide (As2O3) exerts its therapeutic effect by promoting degradation of an oncogenic protein that drives the growth of APL cells, PML-RARalpha (a fusion protein containing sequences from the PML zinc finger protein and retinoic acid receptor alpha). PML and PML-RARalpha degradation is triggered by their SUMOylation, but the mechanism by which As2O3 induces this posttranslational modification is unclear. Here we show that arsenic binds directly to cysteine residues in zinc fingers located within the RBCC domain of PML-RARalpha and PML. Arsenic binding induces PML oligomerization, which increases its interaction with the small ubiquitin-like protein modifier (SUMO)-conjugating enzyme UBC9, resulting in enhanced SUMOylation and degradation. The identification of PML as a direct target of As2O3 provides new insights into the drug's mechanism of action and its specificity for APL.


Asunto(s)
Arsénico/metabolismo , Arsenicales/metabolismo , Arsenicales/farmacología , Proteínas Nucleares/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Óxidos/metabolismo , Óxidos/farmacología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Trióxido de Arsénico , Línea Celular , Humanos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/genética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/genética , Oxazinas/metabolismo , Proteína de la Leucemia Promielocítica , Conformación Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores de Ácido Retinoico/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Receptor alfa de Ácido Retinoico , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Factores de Transcripción/química , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética , Ubiquitinación , Dedos de Zinc
20.
Int J Cancer ; 124(4): 991-7, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19048622

RESUMEN

The major long-term prognostic factor for breast cancer patients treated by first-line chemotherapy is response to treatment. We have previously shown that complete responses to high doses epirubicin-cyclophosphamide were observed only in human tumors bearing a TP53 mutation. Three xenografted human breast tumors, 2 of them with a TP53 mutation and one of them without, were studied for their immediate response to this drug association. Cell cycle, cellular senescence and cell death were characterized and quantified on tissue section before and after treatment. The TP53 wild-type tumor showed a strong early induction of senescence-like phenotype with overexpression of SA-beta-gal and p21(CIP1). In contrast both TP53 mutated tumors showed no sign of cell cycle arrest or senescence. Conversely, abnormal mitoses strongly increased, only in TP53 mutated tumors. Thus, in these in vivo models, epirubicin-cyclophosphamide treatment induces senescence-like features in TP53 wild-type tumor, likely accounting for cell cycle arrest and subsequent resistance to treatment. Conversely in TP53 mutated tumors, chemotherapy induces mitotic catastrophe and tumor death, accounting for complete response to this association exclusively in patients with TP53 mutated tumors.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Genes p53 , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Mutación , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ciclo Celular , Senescencia Celular , Ciclofosfamida/administración & dosificación , Análisis Mutacional de ADN , ADN de Neoplasias , Epirrubicina/administración & dosificación , Humanos , Ratones , Trasplante de Neoplasias , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...