Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Angiogenesis ; 25(2): 205-224, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34665379

RESUMEN

In mice, embryonic dermal lymphatic development is well understood and used to study gene functions in lymphangiogenesis. Notch signaling is an evolutionarily conserved pathway that modulates cell fate decisions, which has been shown to both inhibit and promote dermal lymphangiogenesis. Here, we demonstrate distinct roles for Notch4 signaling versus canonical Notch signaling in embryonic dermal lymphangiogenesis. Actively growing embryonic dermal lymphatics expressed NOTCH1, NOTCH4, and DLL4 which correlated with Notch activity. In lymphatic endothelial cells (LECs), DLL4 activation of Notch induced a subset of Notch effectors and lymphatic genes, which were distinctly regulated by Notch1 and Notch4 activation. Treatment of LECs with VEGF-A or VEGF-C upregulated Dll4 transcripts and differentially and temporally regulated the expression of Notch1 and Hes/Hey genes. Mice nullizygous for Notch4 had an increase in the closure of the lymphangiogenic fronts which correlated with reduced vessel caliber in the maturing lymphatic plexus at E14.5 and reduced branching at E16.5. Activation of Notch4 suppressed LEC migration in a wounding assay significantly more than Notch1, suggesting a dominant role for Notch4 in regulating LEC migration. Unlike Notch4 nulls, inhibition of canonical Notch signaling by expressing a dominant negative form of MAML1 (DNMAML) in Prox1+ LECs led to increased lymphatic density consistent with an increase in LEC proliferation, described for the loss of LEC Notch1. Moreover, loss of Notch4 did not affect LEC canonical Notch signaling. Thus, we propose that Notch4 signaling and canonical Notch signaling have distinct functions in the coordination of embryonic dermal lymphangiogenesis.


Asunto(s)
Linfangiogénesis , Vasos Linfáticos , Animales , Células Endoteliales/metabolismo , Linfangiogénesis/fisiología , Sistema Linfático/metabolismo , Vasos Linfáticos/metabolismo , Ratones , Receptores Notch/metabolismo , Transducción de Señal
2.
Sci Rep ; 14(1): 13603, 2024 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-38866944

RESUMEN

Notch signaling guides vascular development and function by regulating diverse endothelial cell behaviors, including migration, proliferation, vascular density, endothelial junctions, and polarization in response to flow. Notch proteins form transcriptional activation complexes that regulate endothelial gene expression, but few of the downstream effectors that enable these phenotypic changes have been characterized in endothelial cells, limiting our understanding of vascular Notch activities. Using an unbiased screen of translated mRNA rapidly regulated by Notch signaling, we identified novel in vivo targets of Notch signaling in neonatal mouse brain endothelium, including UNC5B, a member of the netrin family of angiogenic-regulatory receptors. Endothelial Notch signaling rapidly upregulates UNC5B in multiple endothelial cell types. Loss or gain of UNC5B recapitulated specific Notch-regulated phenotypes. UNC5B expression inhibited endothelial migration and proliferation and was required for stabilization of endothelial junctions in response to shear stress. Loss of UNC5B partially or wholly blocked the ability of Notch activation to regulate these endothelial cell behaviors. In the developing mouse retina, endothelial-specific loss of UNC5B led to excessive vascularization, including increased vascular outgrowth, density, and branchpoint count. These data indicate that Notch signaling upregulates UNC5B as an effector protein to control specific endothelial cell behaviors and inhibit angiogenic growth.


Asunto(s)
Movimiento Celular , Proliferación Celular , Células Endoteliales , Receptores de Netrina , Receptores Notch , Retina , Transducción de Señal , Animales , Receptores de Netrina/metabolismo , Receptores Notch/metabolismo , Ratones , Células Endoteliales/metabolismo , Retina/metabolismo , Humanos , Vasos Retinianos/metabolismo , Neovascularización Fisiológica
3.
Cancer Res Commun ; 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38984877

RESUMEN

Endothelial Notch signaling is critical for tumor angiogenesis. Notch1 blockade can interfere with tumor vessel function but causes tissue hypoxia and gastrointestinal toxicity. Notch4 is primarily expressed in endothelial cells, where it may promote angiogenesis; however, effective therapeutic targeting of Notch4 has not been successful. We developed highly specific Notch4-blocking antibodies, 6-3-A6 and humanized E7011, allowing therapeutic targeting of Notch4 to be assessed in tumor models. Notch4 was expressed on tumor endothelial cells in multiple cancer models, and endothelial expression was associated with response to E7011/6-3-A6. Anti-Notch4 treatment significantly delayed tumor growth in mouse models of breast, skin, and lung cancer. Enhanced tumor inhibition occurred when anti-Notch4 treatment was used in combination with chemotherapeutics. Endothelial transcriptomic analysis of murine breast tumors treated with 6-3-A6 identified significant changes in pathways of vascular function but caused only modest change in canonical Notch signaling. Analysis of early and late treatment timepoints revealed significant differences in vessel area and perfusion in response to anti-Notch4 treatment. We conclude that targeting Notch4 improves tumor growth control through endothelial intrinsic mechanisms.

4.
Sci Rep ; 12(1): 1655, 2022 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-35102202

RESUMEN

To control sprouting angiogenesis, endothelial Notch signaling suppresses tip cell formation, migration, and proliferation while promoting barrier formation. Each of these responses may be regulated by distinct Notch-regulated effectors. Notch activity is highly dynamic in sprouting endothelial cells, while constitutive Notch signaling drives homeostatic endothelial polarization, indicating the need for both rapid and constitutive Notch targets. In contrast to previous screens that focus on genes regulated by constitutively active Notch, we characterized the dynamic response to Notch. We examined transcriptional changes from 1.5 to 6 h after Notch signal activation via ligand-specific or EGTA induction in cultured primary human endothelial cells and neonatal mouse brain. In each combination of endothelial type and Notch manipulation, transcriptomic analysis identified distinct but overlapping sets of rapidly regulated genes and revealed many novel Notch target genes. Among the novel Notch-regulated signaling pathways identified were effectors in GPCR signaling, notably, the constitutively active GTPase RND1. In endothelial cells, RND1 was shown to be a novel direct Notch transcriptional target and required for Notch control of sprouting angiogenesis, endothelial migration, and Ras activity. We conclude that RND1 is directly regulated by endothelial Notch signaling in a rapid fashion in order to suppress endothelial migration.


Asunto(s)
Encéfalo/irrigación sanguínea , Movimiento Celular , Células Endoteliales/enzimología , Neovascularización Fisiológica , Receptores Notch/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proliferación Celular , Regulación Enzimológica de la Expresión Génica , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Notch/genética , Transducción de Señal , Factores de Tiempo , Transcripción Genética , Proteínas ras/genética , Proteínas ras/metabolismo , Proteínas de Unión al GTP rho/genética
5.
Sci Signal ; 14(679)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33879602

RESUMEN

Chloride intracellular channels 1 (CLIC1) and 4 (CLIC4) are expressed in endothelial cells and regulate angiogenic behaviors in vitro, and the expression of Clic4 is important for vascular development and function in mice. Here, we found that CLIC1 and CLIC4 in endothelial cells regulate critical G protein-coupled receptor (GPCR) pathways associated with vascular development and disease. In cultured endothelial cells, we found that CLIC1 and CLIC4 transiently translocated to the plasma membrane in response to sphingosine 1-phosphate (S1P). Both CLIC1 and CLIC4 were essential for mediating S1P-induced activation of the small guanosine triphosphatase (GTPase) Rac1 downstream of S1P receptor 1 (S1PR1). In contrast, only CLIC1 was essential for S1P-induced activation of the small GTPase RhoA downstream of S1PR2 and S1PR3. Neither were required for other S1P-S1PR signaling outputs. Rescue experiments revealed that CLIC1 and CLIC4 were not functionally interchangeable, suggesting distinct and specific functions for CLICs in transducing GPCR signaling. These CLIC-mediated mechanisms were critical for S1P-induced stimulation of the barrier function in endothelial cell monolayers. Our results define CLICs as previously unknown players in the pathways linking GPCRs to small GTPases and vascular endothelial function.


Asunto(s)
Canales de Cloruro/metabolismo , Proteínas Mitocondriales/metabolismo , Neuropéptidos , Receptores de Esfingosina-1-Fosfato , Proteína de Unión al GTP rac1 , Proteína de Unión al GTP rhoA , Animales , Línea Celular , Células Cultivadas , Células Endoteliales , Lisofosfolípidos , Ratones , Neuropéptidos/metabolismo , Receptores de Lisoesfingolípidos/genética , Transducción de Señal , Esfingosina , Receptores de Esfingosina-1-Fosfato/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
6.
Neurol India ; 57(6): 734-8, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20139501

RESUMEN

BACKGROUND: Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy that affects young boys and the dystrophin gene on the X chromosome has been found to be associated with the disorder. MATERIALS AND METHODS: In this prospective study, 112 clinically diagnosed DMD patients had muscle biopsy and were tested for exon deletions. Genotyping was also carried out at STR44, STR45, STR49 and STR 50 markers in 15 families. RESULTS: Of the 112 clinically suspected DMD patients, the diagnosis of DMD was confirmed by histopathology and/or genetics in 101 patients. The mean age of onset was 3.1+/-1.44 years (1-6 years) and the mean age at presentation was 8.0+/-3.1 years (1.1-18.0 years). Delayed motor milestones were present in 63 (62.3%) patients. The mean creatine kinase value was 11822.64+/-8206.90 U/L (1240-57,700). Eighty-four patients had muscle biopsy and immunohistochemistry was done in 60 muscle samples, all of which demonstrated absence of dystrophin staining. Of the 60 dystrophin-negative cases, 73% showed deletion of at least one exon. Single exon deletion was found in 20.4%. Distal hotspot Exons 45, 47, 49 and 50 were the commonly deleted xenons and the deletion rates were 36%, 35%, 33.7% and 38.5% respectively. CONCLUSIONS: In this study population in south India the deletion rate was 73% and were more frequent in the distal end exon. With the availability of genetic analysis, the first investigation of choice in DMD should be genetic studies and muscle biopsy should be considered only if the genetic tests are negative or not available.


Asunto(s)
Distrofina/genética , Repeticiones de Microsatélite/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Eliminación de Secuencia/genética , Niño , Creatina Quinasa/sangre , Análisis Mutacional de ADN , Distrofina/metabolismo , Exones/genética , Salud de la Familia , Femenino , Genotipo , Humanos , India/epidemiología , Masculino , Relaciones Madre-Hijo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/sangre , Estudios Prospectivos , Sarcoglicanos/metabolismo
7.
Neurol India ; 56(3): 348-51, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18974562

RESUMEN

BACKGROUND: Proximal spinal muscular atrophy (SMA) is a genetically heterogeneous disease with paresis and muscle atrophy due to loss of anterior horn cell function. The survival of motor neuron gene (SMN) and neuronal apoptosis inhibitory protein (NAIP) play a primary role. Both the gene homologues exist as inverted duplications on Chromosome 5q. The telomeric/functional (SMN1) and the centromeric (SMN2) copies differ from each other in eight nucleotides. The C-->T transition (at Codon 280) within Exon 7 of SMN2 causes disruption of an exonic splicing enhancer (ESE) and/or creates an exonic splicing silencer (ESS) leading to abnormal splicing and a truncated protein. OBJECTIVE: To determine the molecular genetics of SMN1 and NAIP genes in SMA from southern India. MATERIALS AND METHODS: In the present study, 37 patients from the neuromuscular disorders clinic of National Institute of Mental Health and Neurosciences were assayed for the deletions in the SMN1 and NAIP genes using PCR-RFLP methods. RESULTS: Among the SMA Type I patients, 43% showed deletions of SMN1 and NAIP. In patients Type II SMA, 57% showed deletions of the SMN1 exons. CONCLUSION: Thus, deletions were found to occur in 47.8% of the Type I and II patients. Lower sensitivity of gene deletion study in clinically suspected SMA needs further study as clinical diagnosis of SMA is not gold standard. However, the results do correlate with other studies conducted in India.


Asunto(s)
Predisposición Genética a la Enfermedad , Atrofia Muscular Espinal/genética , Proteína Inhibidora de la Apoptosis Neuronal/genética , Eliminación de Secuencia/genética , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Adulto , Edad de Inicio , Niño , Análisis Mutacional de ADN , Exones/genética , Genotipo , Humanos , India , Atrofia Muscular Espinal/clasificación , Polimorfismo de Longitud del Fragmento de Restricción/genética
8.
Nat Commun ; 9(1): 1649, 2018 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-29695719

RESUMEN

Recently, we identified ELL2 as a susceptibility gene for multiple myeloma (MM). To understand its mechanism of action, we performed expression quantitative trait locus analysis in CD138+ plasma cells from 1630 MM patients from four populations. We show that the MM risk allele lowers ELL2 expression in these cells (Pcombined = 2.5 × 10-27; ßcombined = -0.24 SD), but not in peripheral blood or other tissues. Consistent with this, several variants representing the MM risk allele map to regulatory genomic regions, and three yield reduced transcriptional activity in plasmocytoma cell lines. One of these (rs3777189-C) co-locates with the best-supported lead variants for ELL2 expression and MM risk, and reduces binding of MAFF/G/K family transcription factors. Moreover, further analysis reveals that the MM risk allele associates with upregulation of gene sets related to ribosome biogenesis, and knockout/knockdown and rescue experiments in plasmocytoma cell lines support a cause-effect relationship. Our results provide mechanistic insight into MM predisposition.


Asunto(s)
Alelos , Cromosomas Humanos Par 5/genética , Regulación Neoplásica de la Expresión Génica , Mieloma Múltiple/genética , Proteínas Ribosómicas/genética , Factores de Elongación Transcripcional/genética , Médula Ósea/patología , Línea Celular Tumoral , Conjuntos de Datos como Asunto , Regulación hacia Abajo , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Predisposición Genética a la Enfermedad , Humanos , Mieloma Múltiple/patología , Células Plasmáticas/metabolismo , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo/genética , Proteínas Ribosómicas/metabolismo , Ribosomas/genética , Ribosomas/metabolismo , Factores de Elongación Transcripcional/metabolismo , Regulación hacia Arriba
9.
Nat Genet ; 49(8): 1182-1191, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28628107

RESUMEN

Immunoglobulins are the effector molecules of the adaptive humoral immune system. In a genome-wide association study of 19,219 individuals, we found 38 new variants and replicated 5 known variants associating with IgA, IgG or IgM levels or with composite immunoglobulin traits, accounted for by 32 loci. Variants at these loci also affect the risk of autoimmune diseases and blood malignancies and influence blood cell development. Notable associations include a rare variant at RUNX3 decreasing IgA levels by shifting isoform proportions (rs188468174[C>T]: P = 8.3 × 10-55, ß = -0.90 s.d.), a rare in-frame deletion in FCGR2B abolishing IgG binding to the encoded receptor (p.Asn106del: P = 4.2 × 10-8, ß = 1.03 s.d.), four IGH locus variants influencing class switching, and ten new associations with the HLA region. Our results provide new insight into the regulation of humoral immunity.


Asunto(s)
Variación Genética , Inmunoglobulinas/genética , Estudios de Cohortes , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Hematopoyesis/genética , Humanos , Islandia , Inmunidad Humoral/genética , Cambio de Clase de Inmunoglobulina/genética , Isotipos de Inmunoglobulinas/genética , Masculino , Polimorfismo de Nucleótido Simple , Suecia
10.
Sci Transl Med ; 8(336): 336ra59, 2016 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-27122612

RESUMEN

Boosting innate immunity represents an important therapeutic alternative to antibiotics. However, the molecular selectivity of this approach is a major concern because innate immune responses often cause collateral tissue damage. We identify the transcription factor interferon regulatory factor 7 (IRF-7), a heterodimer partner of IRF-3, as a target for non-antibiotics-based therapy of bacterial infections. We found that the efficient and self-limiting innate immune response to bacterial infection relies on a tight balance between IRF-3 and IRF-7. Deletion of Irf3 resulted in overexpression of Irf7 and led to an IRF-7-driven hyperinflammatory phenotype, which was entirely prevented if Irf7 was deleted. We then identified a network of strongly up-regulated, IRF-7-dependent genes in Irf3(-/-) mice with kidney pathology, which was absent in Irf7(-/-) mice. IRF-3 and IRF-7 from infected kidney cell nuclear extracts were shown to bind OAS1, CCL5, and IFNB1 promoter oligonucleotides. These data are consistent in children with low IRF7 expression in the blood: attenuating IRF7 promoter polymorphisms (rs3758650-T and rs10902179-G) negatively associated with recurrent pyelonephritis. Finally, we identified IRF-7 as a target for immunomodulatory therapy. Administering liposomal Irf7 siRNA to Irf3(-/-) mice suppressed mucosal IRF-7 expression, and the mice were protected against infection and renal tissue damage. These findings offer a response to the classical but unresolved question of "good versus bad inflammation" and identify IRF7 as a therapeutic target for protection against bacterial infection.


Asunto(s)
Infecciones Bacterianas/inmunología , Inmunidad Innata/fisiología , Factor 7 Regulador del Interferón/metabolismo , Animales , Infecciones Bacterianas/metabolismo , Femenino , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Humanos , Inmunidad Innata/genética , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Riñón/metabolismo , Ratones , Ratones Endogámicos C57BL , Pielonefritis/genética , Pielonefritis/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología
11.
Nat Commun ; 7: 12050, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27363682

RESUMEN

Multiple myeloma (MM) is a plasma cell malignancy with a significant heritable basis. Genome-wide association studies have transformed our understanding of MM predisposition, but individual studies have had limited power to discover risk loci. Here we perform a meta-analysis of these GWAS, add a new GWAS and perform replication analyses resulting in 9,866 cases and 239,188 controls. We confirm all nine known risk loci and discover eight new loci at 6p22.3 (rs34229995, P=1.31 × 10(-8)), 6q21 (rs9372120, P=9.09 × 10(-15)), 7q36.1 (rs7781265, P=9.71 × 10(-9)), 8q24.21 (rs1948915, P=4.20 × 10(-11)), 9p21.3 (rs2811710, P=1.72 × 10(-13)), 10p12.1 (rs2790457, P=1.77 × 10(-8)), 16q23.1 (rs7193541, P=5.00 × 10(-12)) and 20q13.13 (rs6066835, P=1.36 × 10(-13)), which localize in or near to JARID2, ATG5, SMARCD3, CCAT1, CDKN2A, WAC, RFWD3 and PREX1. These findings provide additional support for a polygenic model of MM and insight into the biological basis of tumour development.


Asunto(s)
Mieloma Múltiple/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteína 5 Relacionada con la Autofagia/genética , Estudios de Casos y Controles , Proteínas Cromosómicas no Histona , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Complejo Represivo Polycomb 2/genética , ARN Largo no Codificante/genética , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/genética
12.
Nat Commun ; 6: 7213, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-26007630

RESUMEN

Multiple myeloma (MM) is characterized by an uninhibited, clonal growth of plasma cells. While first-degree relatives of patients with MM show an increased risk of MM, the genetic basis of inherited MM susceptibility is incompletely understood. Here we report a genome-wide association study in the Nordic region identifying a novel MM risk locus at ELL2 (rs56219066T; odds ratio (OR)=1.25; P=9.6 × 10(-10)). This gene encodes a stoichiometrically limiting component of the super-elongation complex that drives secretory-specific immunoglobulin mRNA production and transcriptional regulation in plasma cells. We find that the MM risk allele harbours a Thr298Ala missense variant in an ELL2 domain required for transcription elongation. Consistent with a hypomorphic effect, we find that the MM risk allele also associates with reduced levels of immunoglobulin A (IgA) and G (IgG) in healthy subjects (P=8.6 × 10(-9) and P=6.4 × 10(-3), respectively) and, potentially, with an increased risk of bacterial meningitis (OR=1.30; P=0.0024).


Asunto(s)
Inmunoglobulina A/sangre , Inmunoglobulina G/sangre , Mieloma Múltiple/genética , Proteínas/genética , Factores de Elongación Transcripcional/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Meningitis Bacterianas/genética
13.
PLoS One ; 9(12): e115176, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25503069

RESUMEN

OBJECTIVES: The mechanism by which atheroma plaque becomes unstable is not completely understood to date but analysis of differentially expressed genes in stable versus unstable plaques may provide clues. This will be crucial toward disclosing the mechanistic basis of plaque instability, and may help to identify prognostic biomarkers for ischaemic events. The objective of our study was to identify differences in expression levels of 59 selected genes between symptomatic patients (unstable plaques) and asymptomatic patients (stable plaques). METHODS: 80 carotid plaques obtained by carotid endarterectomy and classified as symptomatic (>70% stenosis) or asymptomatic (>80% stenosis) were used in this study. The expression levels of 59 genes were quantified by qPCR on RNA extracted from the carotid plaques obtained by endarterectomy and analyzed by means of various bioinformatic tools. RESULTS: Several genes associated with autophagy pathways displayed differential expression levels between asymptomatic and symptomatic (i.e. MAP1LC3B, RAB24, EVA1A). In particular, mRNA levels of MAP1LC3B, an autophagic marker, showed a 5-fold decrease in symptomatic samples, which was confirmed in protein blots. Immune system-related factors and endoplasmic reticulum-associated markers (i.e. ERP27, ITPR1, ERO1LB, TIMP1, IL12B) emerged as differently expressed genes between asymptomatic and symptomatic patients. CONCLUSIONS: Carotid atherosclerotic plaques in which MAP1LC3B is underexpressed would not be able to benefit from MAP1LC3B-associated autophagy. This may lead to accumulation of dead cells at lesion site with subsequent plaque destabilization leading to cerebrovascular events. Identified biomarkers and network interactions may represent novel targets for development of treatments against plaque destabilization and thus for the prevention of cerebrovascular events.


Asunto(s)
Enfermedades de las Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/patología , Estenosis Carotídea/metabolismo , Estenosis Carotídea/patología , Proteínas Asociadas a Microtúbulos/biosíntesis , Anciano , Autofagia/fisiología , Biomarcadores , Enfermedades de las Arterias Carótidas/genética , Estenosis Carotídea/genética , Femenino , Expresión Génica , Estudios de Asociación Genética , Humanos , Masculino , Proteínas Asociadas a Microtúbulos/genética , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología
14.
PLoS One ; 8(4): e62376, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23638056

RESUMEN

CD6 has recently been identified and validated as risk gene for multiple sclerosis (MS), based on the association of a single nucleotide polymorphism (SNP), rs17824933, located in intron 1. CD6 is a cell surface scavenger receptor involved in T-cell activation and proliferation, as well as in thymocyte differentiation. In this study, we performed a haptag SNP screen of the CD6 gene locus using a total of thirteen tagging SNPs, of which three were non-synonymous SNPs, and replicated the recently reported GWAS SNP rs650258 in a Spanish-Basque collection of 814 controls and 823 cases. Validation of the six most strongly associated SNPs was performed in an independent collection of 2265 MS patients and 2600 healthy controls. We identified association of haplotypes composed of two non-synonymous SNPs [rs11230563 (R225W) and rs2074225 (A257V)] in the 2(nd) SRCR domain with susceptibility to MS (P max(T) permutation = 1×10(-4)). The effect of these haplotypes on CD6 surface expression and cytokine secretion was also tested. The analysis showed significantly different CD6 expression patterns in the distinct cell subsets, i.e. - CD4(+) naïve cells, P = 0.0001; CD8(+) naïve cells, P<0.0001; CD4(+) and CD8(+) central memory cells, P = 0.01 and 0.05, respectively; and natural killer T (NKT) cells, P = 0.02; with the protective haplotype (RA) showing higher expression of CD6. However, no significant changes were observed in natural killer (NK) cells, effector memory and terminally differentiated effector memory T cells. Our findings reveal that this new MS-associated CD6 risk haplotype significantly modifies expression of CD6 on CD4(+) and CD8(+) T cells.


Asunto(s)
Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/metabolismo , Mapeo Cromosómico , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Adulto , Antígenos CD/química , Antígenos de Diferenciación de Linfocitos T/química , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Análisis por Conglomerados , Citocinas/metabolismo , Femenino , Orden Génico , Sitios Genéticos , Predisposición Genética a la Enfermedad , Haplotipos , Humanos , Desequilibrio de Ligamiento , Activación de Linfocitos/inmunología , Masculino , Polimorfismo de Nucleótido Simple , Dominios y Motivos de Interacción de Proteínas , España , Población Blanca/genética , Adulto Joven
15.
J Neuroimmunol ; 223(1-2): 100-3, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20430450

RESUMEN

A recent meta-analysis of genome-wide association screens coupled to a replication exercise in a combined US/UK collection led to the identification of 4 single nucleotide polymorphisms (SNPs) in three gene loci, i.e. TNFRSF1A, CD6 and IRF8, as novel risk factors for multiple sclerosis with genome-wide level of significance. In the present study, using a combined all-Spain collection of 2515 MS patients and 2942 healthy controls, we demonstrate significant association of rs17824933 in CD6 (P(CMH)=0.004; OR=1.14; 95% CI 1.04-1.24) and of rs1860545 in TNFRSF1A (P(CMH)=0.001; OR=1.15; 95% CI 1.06-1.25) with MS, while the low-frequency coding non-synonymous SNP rs4149584 in TNFRSF1A displayed a trend for association (P(CMH)=0.062; OR=1.27; 95% CI 0.99-1.63). This data reinforce a generic role for CD6 and TNFRSF1A in susceptibility to MS, extending to populations of southern European ancestry.


Asunto(s)
Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos T/genética , Sitios Genéticos/genética , Sitios Genéticos/inmunología , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Adolescente , Adulto , Anciano , Estudios de Casos y Controles , Niño , Estudios de Cohortes , Europa (Continente)/epidemiología , Femenino , Predisposición Genética a la Enfermedad/epidemiología , Predisposición Genética a la Enfermedad/genética , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/epidemiología , Polimorfismo de Nucleótido Simple/inmunología , Factores de Riesgo , España/epidemiología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA