Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Genomics ; 110(2): 98-111, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28911974

RESUMEN

The GLIS family zinc finger 3 isoform (GLIS3) is a risk gene for Type 1 and Type 2 diabetes, glaucoma and Alzheimer's disease endophenotype. We identified GLIS3 binding sites in insulin secreting cells (INS1) (FDR q<0.05; enrichment range 1.40-9.11 fold) sharing the motif wrGTTCCCArTAGs, which were enriched in genes involved in neuronal function and autophagy and in risk genes for metabolic and neuro-behavioural diseases. We confirmed experimentally Glis3-mediated regulation of the expression of genes involved in autophagy and neuron function in INS1 and neuronal PC12 cells. Naturally-occurring coding polymorphisms in Glis3 in the Goto-Kakizaki rat model of type 2 diabetes were associated with increased insulin production in vitro and in vivo, suggestive alteration of autophagy in PC12 and INS1 and abnormal neurogenesis in hippocampus neurons. Our results support biological pleiotropy of GLIS3 in pathologies affecting ß-cells and neurons and underline the existence of trans­nosology pathways in diabetes and its co-morbidities.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Animales , Autofagia , Sitios de Unión , Línea Celular Tumoral , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Hipocampo/citología , Masculino , Neurogénesis , Neuronas/citología , Células PC12 , Polimorfismo Genético , Unión Proteica , Ratas , Ratas Sprague-Dawley , Factores de Transcripción/química , Factores de Transcripción/genética
2.
Blood ; 125(15): 2359-69, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25608561

RESUMEN

The pathogenesis of atypical hemolytic uremic syndrome (aHUS) is strongly linked to dysregulation of the alternative pathway of the complement system. Mutations in complement genes have been identified in about two-thirds of cases, with 5% to 15% being in C3. In this study, 23 aHUS-associated genetic changes in C3 were characterized relative to their interaction with the control proteins factor H (FH), membrane cofactor protein (MCP; CD46), and complement receptor 1 (CR1; CD35). In surface plasmon resonance experiments, 17 mutant recombinant proteins demonstrated a defect in binding to FH and/or MCP, whereas 2 demonstrated reduced binding to CR1. In the majority of cases, decreased binding affinity translated to a decrease in proteolytic inactivation (known as cofactor activity) of C3b via FH and MCP. These results were used to map the putative binding regions of C3b involved in the interaction with MCP and CR1 and interrogated relative to known FH binding sites. Seventy-six percent of patients with C3 mutations had low C3 levels that correlated with disease severity. This study expands our knowledge of the functional consequences of aHUS-associated C3 mutations relative to the interaction of C3 with complement regulatory proteins mediating cofactor activity.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/metabolismo , Complemento C3/genética , Complemento C3/metabolismo , Mapas de Interacción de Proteínas , Síndrome Hemolítico Urémico Atípico/patología , Sitios de Unión , Estudios de Cohortes , Complemento C3/análisis , Femenino , Humanos , Masculino , Modelos Moleculares , Mutación
3.
Biochim Biophys Acta ; 1853(11 Pt A): 2885-96, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26253696

RESUMEN

The Fas associated death domain protein (FADD) is the key adaptor molecule of the apoptotic signal triggered by death receptors of the TNF-R1 superfamily. Besides its crucial role in the apoptotic machinery, FADD has proved to be important in many biological processes like tumorigenesis, embryonic development or cell cycle progression. In a process to decipher the regulatory mechanisms underlying FADD regulation, we identified the anti-apoptotic kinase, CK2, as a new partner and regulator of FADD sub-cellular localization. The blockade of CK2 activity induced FADD re-localization within the cell. Moreover, cytoplasmic FADD was increased when CK2ß was knocked down. In vitro kinase and pull down assays confirmed that FADD could be phosphorylated by the CK2 holoenzyme. We found that phosphorylation is weak with CK2α alone and optimal in the presence of stoichiometric amounts of CK2α catalytic and CK2ß regulatory subunit, showing that FADD phosphorylation is undertaken by the CK2 holoenzyme in a CK2ß-driven fashion. We found that CK2 can phosphorylate FADD on the serine 200 and that this phosphorylation is important for nuclear localization of FADD. Altogether, our results show for the first time that multifaceted kinase, CK2, phosphorylates FADD and is involved in its sub-cellular localization. This work uncovered an important role of CK2 in stable FADD nuclear localization.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Núcleo Celular/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Quinasa de la Caseína II/genética , Línea Celular Tumoral , Núcleo Celular/genética , Proteína de Dominio de Muerte Asociada a Fas/genética , Humanos , Fosforilación/fisiología
4.
J Am Soc Nephrol ; 25(9): 2053-65, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24652797

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is a genetic ultrarare renal disease associated with overactivation of the alternative pathway of complement. Four gain-of-function mutations that form a hyperactive or deregulated C3 convertase have been identified in Factor B (FB) ligand binding sites. Here, we studied the functional consequences of 10 FB genetic changes recently identified from different aHUS cohorts. Using several tests for alternative C3 and C5 convertase formation and regulation, we identified two gain-of-function and potentially disease-relevant mutations that formed either an overactive convertase (M433I) or a convertase resistant to decay by FH (K298Q). One mutation (R178Q) produced a partially cleaved protein with no ligand binding or functional activity. Seven genetic changes led to near-normal or only slightly reduced ligand binding and functional activity compared with the most common polymorphism at position 7, R7. Notably, none of the algorithms used to predict the disease relevance of FB mutations agreed completely with the experimental data, suggesting that in silico approaches should be undertaken with caution. These data, combined with previously published results, suggest that 9 of 15 FB genetic changes identified in patients with aHUS are unrelated to disease pathogenesis. This study highlights that functional assessment of identified nucleotide changes in FB is mandatory to confirm disease association.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/inmunología , Factor B del Complemento/genética , Mutación , Sustitución de Aminoácidos , Sitios de Unión/genética , C3 Convertasa de la Vía Alternativa del Complemento/química , C3 Convertasa de la Vía Alternativa del Complemento/genética , C3 Convertasa de la Vía Alternativa del Complemento/metabolismo , Complemento C3b/metabolismo , C5 Convertasa de la Vía Alternativa del Complemento/química , C5 Convertasa de la Vía Alternativa del Complemento/genética , C5 Convertasa de la Vía Alternativa del Complemento/metabolismo , Factor B del Complemento/química , Factor B del Complemento/metabolismo , Vía Alternativa del Complemento/genética , Simulación por Computador , Frecuencia de los Genes , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ligandos , Modelos Moleculares , Complejos Multiproteicos/química , Polimorfismo Genético , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
5.
Blood ; 119(18): 4182-91, 2012 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-22246034

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is a rare renal thrombotic microangiopathy commonly associated with rare genetic variants in complement system genes, unique to each patient/family. Here, we report 14 sporadic aHUS patients carrying the same mutation, R139W, in the complement C3 gene. The clinical presentation was with a rapid progression to end-stage renal disease (6 of 14) and an unusually high frequency of cardiac (8 of 14) and/or neurologic (5 of 14) events. Although resting glomerular endothelial cells (GEnCs) remained unaffected by R139W-C3 sera, the incubation of those sera with GEnC preactivated with pro-inflammatory stimuli led to increased C3 deposition, C5a release, and procoagulant tissue-factor expression. This functional consequence of R139W-C3 resulted from the formation of a hyperactive C3 convertase. Mutant C3 showed an increased affinity for factor B and a reduced binding to membrane cofactor protein (MCP; CD46), but a normal regulation by factor H (FH). In addition, the frequency of at-risk FH and MCP haplotypes was significantly higher in the R139W-aHUS patients, compared with normal donors or to healthy carriers. These genetic background differences could explain the R139W-aHUS incomplete penetrance. These results demonstrate that this C3 mutation, especially when associated with an at-risk FH and/or MCP haplotypes, becomes pathogenic following an inflammatory endothelium-damaging event.


Asunto(s)
Complemento C3/genética , Síndrome Hemolítico-Urémico/genética , Mutación Missense , Mutación Puntual , Adolescente , Adulto , Anciano , Sustitución de Aminoácidos , Síndrome Hemolítico Urémico Atípico , Células Cultivadas/efectos de los fármacos , Preescolar , Complemento C3/química , Complemento C3/metabolismo , Factor B del Complemento/metabolismo , Progresión de la Enfermedad , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Haplotipos/genética , Síndrome Hemolítico-Urémico/sangre , Síndrome Hemolítico-Urémico/complicaciones , Síndrome Hemolítico-Urémico/inmunología , Humanos , Lactante , Fallo Renal Crónico/etiología , Glomérulos Renales/patología , Masculino , Proteína Cofactora de Membrana/metabolismo , Persona de Mediana Edad , Modelos Moleculares , Penetrancia , Conformación Proteica , Resonancia por Plasmón de Superficie , Adulto Joven
6.
Nephrol Dial Transplant ; 28(11): 2899-907, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24009284

RESUMEN

BACKGROUND: Atypical haemolytic uraemic syndrome (aHUS) is associated with dysfunction of the alternative pathway of complement. Disease activity subsides as renal failure progresses but recurs upon renal transplantation, indicating that viable renal tissue contributes to disease activity. We present evidence of cerebrovascular occlusive disease indicating that vascular injury may occur in the absence of kidneys. METHODS: A currently 12-year-old girl developed renal failure at the age of 20 months. She underwent bilateral nephrectomy and renal transplantation but lost the transplant due to recurrences. She was on haemodialysis for 7 years. At 10 years of age she developed a transient ischaemic attack. Imaging, genetic investigation and mutation characterization were performed. RESULTS: Imaging demonstrated occlusion and stenosis of the carotid arteries. Two complement mutations, a novel mutation in factor B and a previously described mutation in factor I, and the H3-factor H haplotype, were identified. The factor B mutation, L433S, did not induce excessive complement activation in vitro. Measurement of C3 degradation products indicated ongoing complement activation. In spite of the patient being anephric, treatment was initiated with eculizumab, a humanized anti-C5 antibody that blocks terminal complement activation. She underwent a successful kidney transplant 9 months later and has not developed a recurrence or progression of vascular stenosis 1 year later. CONCLUSIONS: The course of disease in this patient with aHUS suggests that complement-mediated vascular injury may occur in the total absence of renal tissue and overt recurrences. To our knowledge, this is the first description of eculizumab treatment in an anephric aHUS patient.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Trastornos Cerebrovasculares/tratamiento farmacológico , Síndrome Hemolítico-Urémico/tratamiento farmacológico , Trasplante de Riñón , Nefrectomía/efectos adversos , Insuficiencia Renal/complicaciones , Síndrome Hemolítico Urémico Atípico , Trastornos Cerebrovasculares/etiología , Trastornos Cerebrovasculares/patología , Niño , Complemento C5/metabolismo , Factor B del Complemento/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Síndrome Hemolítico-Urémico/etiología , Síndrome Hemolítico-Urémico/patología , Humanos , Imagen por Resonancia Magnética , Mutación/genética , Reacción en Cadena de la Polimerasa , Pronóstico , Insuficiencia Renal/cirugía , Resonancia por Plasmón de Superficie
7.
Elife ; 122023 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-36848406

RESUMEN

Bone destruction is a hallmark of chronic inflammation, and bone-resorbing osteoclasts arising under such a condition differ from steady-state ones. However, osteoclast diversity remains poorly explored. Here, we combined transcriptomic profiling, differentiation assays and in vivo analysis in mouse to decipher specific traits for inflammatory and steady-state osteoclasts. We identified and validated the pattern-recognition receptors (PRR) Tlr2, Dectin-1, and Mincle, all involved in yeast recognition as major regulators of inflammatory osteoclasts. We showed that administration of the yeast probiotic Saccharomyces boulardii CNCM I-745 (Sb) in vivo reduced bone loss in ovariectomized but not sham mice by reducing inflammatory osteoclastogenesis. This beneficial impact of Sb is mediated by the regulation of the inflammatory environment required for the generation of inflammatory osteoclasts. We also showed that Sb derivatives as well as agonists of Tlr2, Dectin-1, and Mincle specifically inhibited directly the differentiation of inflammatory but not steady-state osteoclasts in vitro. These findings demonstrate a preferential use of the PRR-associated costimulatory differentiation pathway by inflammatory osteoclasts, thus enabling their specific inhibition, which opens new therapeutic perspectives for inflammatory bone loss.


Asunto(s)
Osteoporosis , Probióticos , Animales , Ratones , Osteogénesis , Osteoporosis/terapia , Receptor Toll-Like 2 , Saccharomyces/genética , Saccharomyces/metabolismo
8.
Joint Bone Spine ; 89(6): 105419, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35640836

RESUMEN

OBJECTIVES: The strong heritability of spondyloarthritis remains poorly explained, despite several large-scale association studies. A recent linkage analysis identified a new region linked to SpA on 13q13. Here we searched for variants potentially explaining this linkage signal by deep-sequencing of the region. METHODS: Re-sequencing of the 1.4 Mb target interval was performed in 92 subjects from the 43 best-linked multicases families (71 spondyloarthritis and 21 unaffected relatives), using hybridization capture-based protocol (Illumina Nextera®). Variants of interest were then genotyped by TaqMan and high resolution melting to check their co-segregation with disease in the same families and to test their association with spondyloarthritis in an independent cohort of 1,091 unrelated cases and 399 controls. Expression of FREM2 was assessed by immunostaining. RESULTS: Of the 7,563 variants identified, 24 were non-synonymous coding single-nucleotide variants. Two of them were located in the FREM2 gene on a haplotype co-segregating with the disease, including one common variant (R1840W, minor allele frequency=0.11) and one rare variant (R727H, minor allele frequency=0.0001). In the case-control analysis, there was no significant association between R1840W and spondyloarthritis (P-value=0.21), whereas R727H was not detected in any of the genotyped individuals. Immunostaining experiments revealed that FREM2 is expressed in synovial membrane, cartilage and colon. CONCLUSIONS: Targeted re-sequencing of a spondyloarthritis-linked region allowed us to identify a rare non-synonymous coding variant in FREM2, co-segregating with spondyloarthritis in a large family. This gene is expressed in several tissues relevant to spondyloarthritis pathogenesis, supporting its putative implication in spondyloarthritis.


Asunto(s)
Predisposición Genética a la Enfermedad , Espondiloartritis , Humanos , Polimorfismo de Nucleótido Simple , Ligamiento Genético , Espondiloartritis/genética , Genotipo , Proteínas de la Matriz Extracelular/genética
9.
Blood ; 114(13): 2837-45, 2009 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-19584399

RESUMEN

Complement is a major innate immune defense against pathogens, tightly regulated to prevent host tissue damage. Atypical hemolytic uremic syndrome (aHUS) is characterized by endothelial damage leading to renal failure and is highly associated with abnormal alternative pathway regulation. We characterized the functional consequences of 2 aHUS-associated mutations (D(254)G and K(325)N) in factor B, a key participant in the alternative C3 convertase. Mutant proteins formed high-affinity C3-binding site, leading to a hyperfunctional C3 convertase, resistant to decay by factor H. This led to enhanced complement deposition on the surface of alternative pathway activator cells. In contrast to native factor B, the 2 mutants bound to inactivated C3 and induced formation of functional C3-convertase on iC3b-coated surface. We demonstrated for the first time that factor B mutations lead to enhanced C3-fragment deposition on quiescent and adherent human glomerular cells (GEnCs) and human umbilical vein endothelial cells (HUVECs), together with the formation of sC5b-9 complexes. These results could explain the occurrence of the disease, since excessive complement deposition on endothelial cells is a central event in the pathogenesis of aHUS. Therefore, risk factors for aHUS are not only mutations leading to loss of regulation, but also mutations, resulting in hyperactive C3 convertase.


Asunto(s)
Convertasas de Complemento C3-C5/fisiología , Proteínas del Sistema Complemento/metabolismo , Células Endoteliales/metabolismo , Síndrome Hemolítico-Urémico/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Células Cultivadas , Niño , Preescolar , Estudios de Cohortes , Activación de Complemento/genética , Convertasas de Complemento C3-C5/genética , Proteínas del Sistema Complemento/genética , Familia , Femenino , Síndrome Hemolítico-Urémico/inmunología , Síndrome Hemolítico-Urémico/metabolismo , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Modelos Moleculares , Proteínas Mutantes/fisiología , Linaje , Adulto Joven
10.
Arthritis Res Ther ; 23(1): 246, 2021 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-34560894

RESUMEN

BACKGROUND: In patients with axial spondyloarthritis (axSpA), monocytes show a pre-activated phenotype. Gut inflammation is a trigger of monocyte activation and may also affect their development in the bone marrow (BM). As gut inflammation is commonly observed in axSpA patients, we performed a detailed analysis of monocyte transcriptomes of axSpA patients in two cohorts and searched for signs of activation and developmental adaptations as putative imprints of gut inflammation. METHODS: Transcriptomes of blood CD14+ monocytes of HLA-B27+ axSpA patients and healthy controls (HC) were generated by microarrays from cohort 1 and by RNA-sequencing from cohort 2. Differentially expressed genes from both analyses were subjected to gene set enrichment analysis (GSEA) and to co-expression analysis in reference transcriptomes from BM cells, blood cells and activated monocytes. As serological markers of translocation, 1,3 beta-glycan, intestinal fatty acid binding protein, and lipopolysaccharide binding protein (LBP) were determined by LAL and ELISA. RESULTS: Transcriptome analysis identified axSpA-specific monocyte signatures showing an imprint of LPS/cytokine-activated monocytes, late granulopoietic BM cells, blood neutrophils, and G-CSF-mobilized blood cells, which suggests LPS/TNF activation and more prominent BM adaptation promoting a neutrophil-like phenotype. GSEA mapped axSpA upregulated genes to inflammatory responses and TNFα signaling and downregulated probe-sets to metabolic pathways. Among translocation markers, LBP levels were significantly increased in axSpA patients vs. HC (p < 0.001). Stratified analysis by disease activity and stage identified an "active disease signature" (BASDAI ≥ 4) with an imprint of LPS/cytokine-activated monocytes and CD16+ monocyte subsets. The "AS signature" (vs. non-radiographic axSpA) showed a reinforced neutrophil-like phenotype due to deprivation of dendritic cell transcripts. CONCLUSIONS: The neutrophil-like phenotype of axSpA monocytes points towards a biased monocytopoiesis from granulocyte-monocyte progenitors. This shift in monocytopoiesis and the LPS/cytokine imprint as well as the elevated LBP levels are indicators of systemic inflammation, which may result from bacterial translocation. The BM adaptation is most prominent in AS patients while disease activity appears to be linked to activation and trafficking of monocytes.


Asunto(s)
Monocitos , Espondiloartritis , Citocinas , Perfilación de la Expresión Génica , Humanos , Espondiloartritis/genética , Transcriptoma
11.
J Clin Invest ; 117(8): 2225-32, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17671652

RESUMEN

Recent reports have challenged the notion that retroviruses and retroviral vectors integrate randomly into the host genome. These reports pointed to a strong bias toward integration in and near gene coding regions and, for gammaretroviral vectors, around transcription start sites. Here, we report the results obtained from a large-scale mapping of 572 retroviral integration sites (RISs) isolated from cells of 9 patients with X-linked SCID (SCID-X1) treated with a retrovirus-based gene therapy protocol. Our data showed that two-thirds of insertions occurred in or very near to genes, of which more than half were highly expressed in CD34(+) progenitor cells. Strikingly, one-fourth of all integrations were clustered as common integration sites (CISs). The highly significant incidence of CISs in circulating T cells and the nature of their locations indicate that insertion in many gene loci has an influence on cell engraftment, survival, and proliferation. Beyond the observed cases of insertional mutagenesis in 3 patients, these data help to elucidate the relationship between vector insertion and long-term in vivo selection of transduced cells in human patients with SCID-X1.


Asunto(s)
Gammaretrovirus , Terapia Genética , Vectores Genéticos , Genoma Humano , Linfopoyesis/genética , Integración Viral/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Antígenos CD34 , Proliferación Celular , Supervivencia Celular/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Mutagénesis Insercional , Sitios de Carácter Cuantitativo , Linfocitos T , Factores de Tiempo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética
12.
Joint Bone Spine ; 87(6): 565-571, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32534204

RESUMEN

Spondyloarthritis (SpA) is a chronic inflammatory disorder resulting from a combination of genetic predisposition and environmental factors. Despite recent advances, a substantial fraction of its genetic basis remains poorly understood. Several mechanisms have been proposed to account for this unexplained heritability, including epigenetics which can play a role at the interface between genetic and environmental susceptibility factors. Epigenetics refers to changes in gene expression that are not encoded in the DNA sequence itself. Such mechanisms may include DNA methylation, histone modifications and non-coding RNAs. Disruption of one of these systems can lead to inappropriate gene expression, which in turn might favour the development of disease. Thanks to recent technological progress, there has been a growing interest in the field of epigenetics in complex diseases, including SpA. However, epigenetic studies face some methodological limitations that hamper interpretation of their results: small sample size, absence of biological replication, lack of adequate controls for potential confounders, studies not performed in the most relevant cell/tissues. In the future, integration of epigenetics with other "omics" data will probably be necessary to improve our understanding of SpA pathogenesis. These issues need to be addressed before considering the use of epigenetic marks in clinical routine, as biomarkers or as drug targets.


Asunto(s)
Epigénesis Genética , Espondiloartritis , Metilación de ADN , Predisposición Genética a la Enfermedad , Humanos , Espondiloartritis/genética
13.
Elife ; 92020 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-32400390

RESUMEN

Bone destruction relies on interactions between bone and immune cells. Bone-resorbing osteoclasts (OCLs) were recently identified as innate immune cells activating T cells toward tolerance or inflammation. Thus, pathological bone destruction not only relies on increased osteoclast differentiation, but also on the presence of inflammatory OCLs (i-OCLs), part of which express Cx3cr1. Here, we investigated the contribution of mouse Cx3cr1+ and Cx3cr1neg i-OCLs to bone loss. We showed that Cx3cr1+ and Cx3cr1neg i-OCLs differ considerably in transcriptional and functional aspects. Cx3cr1neg i-OCLs have a high ability to resorb bone and activate inflammatory CD4+ T cells. Although Cx3cr1+ i-OCLs are associated with inflammation, they resorb less and have in vitro an immune-suppressive effect on Cx3cr1neg i-OCLs, mediated by PD-L1. Our results provide new insights into i-OCL heterogeneity. They also reveal that different i-OCL subsets may interact to regulate inflammation. This contributes to a better understanding and prevention of inflammatory bone destruction.


Asunto(s)
Resorción Ósea/metabolismo , Receptor 1 de Quimiocinas CX3C/metabolismo , Inflamación/metabolismo , Osteoclastos/metabolismo , Osteogénesis , Osteoporosis/metabolismo , Animales , Resorción Ósea/inmunología , Resorción Ósea/patología , Resorción Ósea/prevención & control , Receptor 1 de Quimiocinas CX3C/genética , Comunicación Celular , Células Cultivadas , Femenino , Inflamación/inmunología , Inflamación/patología , Inflamación/prevención & control , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoclastos/inmunología , Osteoclastos/patología , Osteoporosis/inmunología , Osteoporosis/patología , Osteoporosis/prevención & control , Ovariectomía , Fenotipo , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
14.
Mol Ther ; 16(2): 396-403, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18223550

RESUMEN

Severe combined immunodeficiency (SCID) caused by mutation of the recombination-activating gene 1 (RAG1) or Artemis gene lead to the absence of B- and T-cell differentiation. The only curative treatment is allogeneic bone marrow (BM) transplantation, which displays a high survival rate when an HLA compatible donor is available but has a poorer prognosis when the donor is partially compatible. Consequently, gene therapy may be a promising alternative strategy for these diseases. Here, we report that lentiviral gene-corrected BM CD34(+) cells (isolated from Artemis- or RAG1-deficient patients) sustain human B-cell differentiation following injection into non-obese diabetic/SCID (NOD-SCID) mice previously infused with anti-interleukin-2 receptor beta chain monoclonal antibody. In most of the mice BM, engrafted with Artemis-transduced cells, human B-cell differentiation occurred until the mature stage. The B cells were functional as human immunoglobulin M (IgM) was present in the serum. Following injection with RAG1-transduced cells, human engraftment occurred in vivo but B-cell differentiation until the mature stage was less frequent. However, when it occurred, it was always associated with human IgM production. This overall approach represents a useful tool for evaluating gene transfer efficiency in human SCID forms affecting B-cell development (such as Artemis deficiency) and for testing new vectors for improving in vivo RAG1 complementation.


Asunto(s)
Antígenos CD34/fisiología , Linfocitos B/citología , Trasplante de Médula Ósea/métodos , Diferenciación Celular/fisiología , Animales , Anticuerpos Monoclonales/inmunología , Antígenos CD34/genética , Linfocitos B/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/genética , Proteínas de Unión al ADN , Endonucleasas , Proteínas de Homeodominio/genética , Humanos , Inmunoglobulina M/metabolismo , Subunidad beta del Receptor de Interleucina-2/inmunología , Lentivirus/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Nucleares/genética
15.
N Engl J Med ; 346(16): 1185-93, 2002 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-11961146

RESUMEN

BACKGROUND: X-linked severe combined immunodeficiency due to a mutation in the gene encoding the common gamma (gamma(c)) chain is a lethal condition that can be cured by allogeneic stem-cell transplantation. We investigated whether infusion of autologous hematopoietic stem cells that had been transduced in vitro with the gamma(c) gene can restore the immune system in patients with severe combined immunodeficiency. METHODS: CD34+ bone marrow cells from five boys with X-linked severe combined immunodeficiency were transduced ex vivo with the use of a defective retroviral vector. Integration and expression of the gamma(c) transgene and development of lymphocyte subgroups and their functions were sequentially analyzed over a period of up to 2.5 years after gene transfer. RESULTS: No adverse effects resulted from the procedure. Transduced T cells and natural killer cells appeared in the blood of four of the five patients within four months. The numbers and phenotypes of T cells, the repertoire of T-cell receptors, and the in vitro proliferative responses of T cells to several antigens after immunization were nearly normal up to two years after treatment. Thymopoiesis was documented by the presence of naive T cells and T-cell antigen-receptor episomes and the development of a normal-sized thymus gland. The frequency of transduced B cells was low, but serum immunoglobulin levels and antibody production after immunization were sufficient to avoid the need for intravenous immunoglobulin. Correction of the immunodeficiency eradicated established infections and allowed patients to have a normal life. CONCLUSIONS: Ex vivo gene therapy with gamma(c) can safely correct the immune deficiency of patients with X-linked severe combined immunodeficiency.


Asunto(s)
Terapia Genética , Inmunodeficiencia Combinada Grave/terapia , Anticuerpos/sangre , Antígenos CD/análisis , Células de la Médula Ósea , Ligamiento Genético , Vectores Genéticos , Humanos , Inmunoglobulinas/sangre , Inmunoglobulinas Intravenosas , Lactante , Células Asesinas Naturales/fisiología , Recuento de Linfocitos , Masculino , Virus de la Leucemia Murina de Moloney/genética , Receptores de Citocinas/genética , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Linfocitos T/inmunología , Linfocitos T/fisiología , Transducción Genética , Transgenes , Cromosoma X
16.
Cell Rep ; 20(1): 136-148, 2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-28683308

RESUMEN

The influence of the gut microbiome on metabolic and behavioral traits is widely accepted, though the microbiome-derived metabolites involved remain unclear. We carried out untargeted urine 1H-NMR spectroscopy-based metabolic phenotyping in an isogenic C57BL/6J mouse population (n = 50) and show that microbial-host co-metabolites are prodromal (i.e., early) markers predicting future divergence in metabolic (obesity and glucose homeostasis) and behavioral (anxiety and activity) outcomes with 94%-100% accuracy. Some of these metabolites also modulate disease phenotypes, best illustrated by trimethylamine-N-oxide (TMAO), a product of microbial-host co-metabolism predicting future obesity, impaired glucose tolerance (IGT), and behavior while reducing endoplasmic reticulum stress and lipogenesis in 3T3-L1 adipocytes. Chronic in vivo TMAO treatment limits IGT in HFD-fed mice and isolated pancreatic islets by increasing insulin secretion. We highlight the prodromal potential of microbial metabolites to predict disease outcomes and their potential in shaping mammalian phenotypic heterogeneity.


Asunto(s)
Ansiedad/microbiología , Microbioma Gastrointestinal , Intolerancia a la Glucosa/microbiología , Metaboloma , Obesidad/microbiología , Fenotipo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Ansiedad/metabolismo , Biomarcadores/metabolismo , Glucemia/metabolismo , Línea Celular , Estrés del Retículo Endoplásmico , Intolerancia a la Glucosa/metabolismo , Interacciones Huésped-Patógeno , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Lipogénesis , Masculino , Metilaminas/farmacología , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Oxidantes/farmacología
17.
G3 (Bethesda) ; 6(11): 3671-3683, 2016 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-27646706

RESUMEN

To test the impact of genetic heterogeneity on cis- and trans-mediated mechanisms of gene expression regulation, we profiled the transcriptome of adipose tissue in 20 inbred congenic strains derived from diabetic Goto-Kakizaki (GK) rats and Brown-Norway (BN) controls, which contain well-defined blocks (1-183 Mb) of genetic polymorphisms, and in 123 genetically heterogeneous rats of an (GK × BN)F2 offspring. Within each congenic we identified 73-1351 differentially expressed genes (DEGs), only 7.7% of which mapped within the congenic blocks, and which may be regulated in cis The remainder localized outside the blocks, and therefore must be regulated in trans Most trans-regulated genes exhibited approximately twofold expression changes, consistent with monoallelic expression. Altered biological pathways were replicated between congenic strains sharing blocks of genetic polymorphisms, but polymorphisms at different loci also had redundant effects on transcription of common distant genes and pathways. We mapped 2735 expression quantitative trait loci (eQTL) in the F2 cross, including 26% predominantly cis-regulated genes, which validated DEGs in congenic strains. A hotspot of >300 eQTL in a 10 cM region of chromosome 1 was enriched in DEGs in a congenic strain. However, many DEGs among GK, BN and congenic strains did not replicate as eQTL in F2 hybrids, demonstrating distinct mechanisms of gene expression when alleles segregate in an outbred population or are fixed homozygous across the entire genome or in short genomic regions. Our analysis provides conceptual advances in our understanding of the complex architecture of genome expression and pathway regulation, and suggests a prominent impact of epistasis and monoallelic expression on gene transcription.

18.
Genome Med ; 8(1): 101, 2016 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-27716393

RESUMEN

BACKGROUND: The genetic regulation of metabolic phenotypes (i.e., metabotypes) in type 2 diabetes mellitus occurs through complex organ-specific cellular mechanisms and networks contributing to impaired insulin secretion and insulin resistance. Genome-wide gene expression profiling systems can dissect the genetic contributions to metabolome and transcriptome regulations. The integrative analysis of multiple gene expression traits and metabolic phenotypes (i.e., metabotypes) together with their underlying genetic regulation remains a challenge. Here, we introduce a systems genetics approach based on the topological analysis of a combined molecular network made of genes and metabolites identified through expression and metabotype quantitative trait locus mapping (i.e., eQTL and mQTL) to prioritise biological characterisation of candidate genes and traits. METHODS: We used systematic metabotyping by 1H NMR spectroscopy and genome-wide gene expression in white adipose tissue to map molecular phenotypes to genomic blocks associated with obesity and insulin secretion in a series of rat congenic strains derived from spontaneously diabetic Goto-Kakizaki (GK) and normoglycemic Brown-Norway (BN) rats. We implemented a network biology strategy approach to visualize the shortest paths between metabolites and genes significantly associated with each genomic block. RESULTS: Despite strong genomic similarities (95-99 %) among congenics, each strain exhibited specific patterns of gene expression and metabotypes, reflecting the metabolic consequences of series of linked genetic polymorphisms in the congenic intervals. We subsequently used the congenic panel to map quantitative trait loci underlying specific mQTLs and genome-wide eQTLs. Variation in key metabolites like glucose, succinate, lactate, or 3-hydroxybutyrate and second messenger precursors like inositol was associated with several independent genomic intervals, indicating functional redundancy in these regions. To navigate through the complexity of these association networks we mapped candidate genes and metabolites onto metabolic pathways and implemented a shortest path strategy to highlight potential mechanistic links between metabolites and transcripts at colocalized mQTLs and eQTLs. Minimizing the shortest path length drove prioritization of biological validations by gene silencing. CONCLUSIONS: These results underline the importance of network-based integration of multilevel systems genetics datasets to improve understanding of the genetic architecture of metabotype and transcriptomic regulation and to characterize novel functional roles for genes determining tissue-specific metabolism.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Metaboloma , Sitios de Carácter Cuantitativo , Carácter Cuantitativo Heredable , Transcriptoma , Animales , Animales Congénicos , Mapeo Cromosómico , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ontología de Genes , Redes Reguladoras de Genes , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Masculino , Redes y Vías Metabólicas , Anotación de Secuencia Molecular , Ratas Endogámicas BN , Biología de Sistemas
19.
PLoS One ; 9(3): e91375, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24675842

RESUMEN

MicroRNAs are emerging as new mediators in the regulation of adipose tissue biology and the development of obesity. An important role of microRNA-125a has been suggested in the pathogenesis of insulin resistance (IR). Here, we characterized the function of microRNA-125a in adipose tissue in a context of experimentally-induced IR and obesity in mice and in obese patients. We showed time dependent overexpression of the microRNA in adipose tissue of BALB/c and C57BL/6J mice in response to high fat diet (HFD) feeding. MicroRNA-125a expression was downregulated in vitro in insulin resistant 3T3-L1 adipocytes and ex vivo in adipose tissue of obese patients. In vitro modulation of microRNA-125a expression in 3T3-L1 adipocytes did not affect glucose uptake. Gene set enrichment analysis (GSEA) identified significantly altered expression patterns of predicted microRNA-125a gene targets in transcriptomic datasets of adipose tissue from HFD-fed mice and obese patients. Among genes that contributed to global enrichment of altered expression of microRNA-125a targets, Thyrotroph embryonic factor (Tef), Mannan-binding lectin serine peptidase 1, Reticulon 2 and Ubiquitin-conjugating enzyme E2L3 were significantly differentially expressed in adipose tissue in these groups. We showed that Tef expression is reduced in adipose tissue of obese patients following gastric bypass surgery. Our findings indicate that microRNA-125a expression in adipose tissue adapts to IR and may play a role in the development of obesity in mice and obese subjects through uncoupled regulation of the expression of microRNA-125a and its targets.


Asunto(s)
Adaptación Biológica/genética , Tejido Adiposo/metabolismo , Regulación de la Expresión Génica , MicroARNs/genética , Obesidad/genética , Células 3T3-L1 , Adipocitos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Cirugía Bariátrica , Biomarcadores , Diferenciación Celular , Análisis por Conglomerados , Dieta Alta en Grasa , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Glucosa/metabolismo , Humanos , Resistencia a la Insulina , Masculino , Ratones , Persona de Mediana Edad , Obesidad/diagnóstico , Obesidad/metabolismo , Obesidad/cirugía , Fenotipo , Interferencia de ARN , Factores Sexuales , Transcriptoma
20.
PLoS One ; 8(12): e82825, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24324835

RESUMEN

Nutritional factors play important roles in the etiology of obesity, type 2 diabetes mellitus and their complications through genotype x environment interactions. We have characterised molecular adaptation to high fat diet (HFD) feeding in inbred mouse strains widely used in genetic and physiological studies. We carried out physiological tests, plasma lipid assays, obesity measures, liver histology, hepatic lipid measurements and liver genome-wide gene transcription profiling in C57BL/6J and BALB/c mice fed either a control or a high fat diet. The two strains showed marked susceptibility (C57BL/6J) and relative resistance (BALB/c) to HFD-induced insulin resistance and non alcoholic fatty liver disease (NAFLD). Global gene set enrichment analysis (GSEA) of transcriptome data identified consistent patterns of expression of key genes (Srebf1, Stard4, Pnpla2, Ccnd1) and molecular pathways in the two strains, which may underlie homeostatic adaptations to dietary fat. Differential regulation of pathways, including the proteasome, the ubiquitin mediated proteolysis and PPAR signalling in fat fed C57BL/6J and BALB/c suggests that altered expression of underlying diet-responsive genes may be involved in contrasting nutrigenomic predisposition and resistance to insulin resistance and NAFLD in these models. Collectively, these data, which further demonstrate the impact of gene x environment interactions on gene expression regulations, contribute to improved knowledge of natural and pathogenic adaptive genomic regulations and molecular mechanisms associated with genetically determined susceptibility and resistance to metabolic diseases.


Asunto(s)
Dieta Alta en Grasa , Hígado Graso/etiología , Hígado Graso/metabolismo , Nutrigenómica , Obesidad/complicaciones , Obesidad/etiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Adaptación Fisiológica , Animales , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Glucosa/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Enfermedad del Hígado Graso no Alcohólico , Reproducibilidad de los Resultados , Transducción de Señal , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA