Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Ann Rheum Dis ; 76(2): 468-472, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27566796

RESUMO

OBJECTIVES: Familial chilblain lupus is a monogenic form of cutaneous lupus erythematosus caused by loss-of-function mutations in the nucleases TREX1 or SAMHD1. In a family without TREX1 or SAMHD1 mutation, we sought to determine the causative gene and the underlying disease pathology. METHODS: Exome sequencing was used for disease gene identification. Structural analysis was performed by homology modelling and docking simulations. Type I interferon (IFN) activation was assessed in cells transfected with STING cDNA using an IFN-ß reporter and Western blotting. IFN signatures in patient blood in response to tofacitinib treatment were measured by RT-PCR of IFN-stimulated genes. RESULTS: In a multigenerational family with five members affected with chilblain lupus, we identified a heterozygous mutation of STING, a signalling molecule in the cytosolic DNA sensing pathway. Structural and functional analyses indicate that mutant STING enhances homodimerisation in the absence of its ligand cGAMP resulting in constitutive type I IFN activation. Treatment of two affected family members with the Janus kinase (JAK) inhibitor tofacitinib led to a marked suppression of the IFN signature. CONCLUSIONS: A heterozygous gain-of-function mutation in STING can cause familial chilblain lupus. These findings expand the genetic spectrum of type I IFN-dependent disorders and suggest that JAK inhibition may be of therapeutic value.


Assuntos
Pérnio/genética , Lúpus Eritematoso Cutâneo/genética , Proteínas de Membrana/genética , Adulto , Western Blotting , Pérnio/tratamento farmacológico , Pérnio/imunologia , Pérnio/patologia , Família , Feminino , Grécia , Humanos , Interferon Tipo I/imunologia , Interferon beta/imunologia , Lúpus Eritematoso Cutâneo/tratamento farmacológico , Lúpus Eritematoso Cutâneo/imunologia , Lúpus Eritematoso Cutâneo/patologia , Masculino , Angioscopia Microscópica , Simulação de Acoplamento Molecular , Mutação , Linhagem , Piperidinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/uso terapêutico , Pirróis/uso terapêutico , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pele/patologia
2.
Ann Rheum Dis ; 74(3): e17, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24445253

RESUMO

OBJECTIVES: The HIV restriction factor, SAMHD1 (SAM domain and HD domain-containing protein 1), is a triphosphohydrolase that degrades deoxyribonucleoside triphosphates (dNTPs). Mutations in SAMHD1 cause Aicardi-Goutières syndrome (AGS), an inflammatory disorder that shares phenotypic similarity with systemic lupus erythematosus, including activation of antiviral type 1 interferon (IFN). To further define the pathomechanisms underlying autoimmunity in AGS due to SAMHD1 mutations, we investigated the physiological properties of SAMHD1. METHODS: Primary patient fibroblasts were examined for dNTP levels, proliferation, senescence, cell cycle progression and DNA damage. Genome-wide transcriptional profiles were generated by RNA sequencing. Interaction of SAMHD1 with cyclin A was assessed by coimmunoprecipitation and fluorescence cross-correlation spectroscopy. Cell cycle-dependent phosphorylation of SAMHD1 was examined in synchronised HeLa cells and using recombinant SAMHD1. SAMHD1 was knocked down by RNA interference. RESULTS: We show that increased dNTP pools due to SAMHD1 deficiency cause genome instability in fibroblasts of patients with AGS. Constitutive DNA damage signalling is associated with cell cycle delay, cellular senescence, and upregulation of IFN-stimulated genes. SAMHD1 is phosphorylated by cyclin A/cyclin-dependent kinase 1 in a cell cycle-dependent manner, and its level fluctuates during the cell cycle, with the lowest levels observed in G1/S phase. Knockdown of SAMHD1 by RNA interference recapitulates activation of DNA damage signalling and type 1 IFN activation. CONCLUSIONS: SAMHD1 is required for genome integrity by maintaining balanced dNTP pools. dNTP imbalances due to SAMHD1 deficiency cause DNA damage, leading to intrinsic activation of IFN signalling. These findings establish a novel link between DNA damage signalling and innate immune activation in the pathogenesis of autoimmunity.


Assuntos
Doenças Autoimunes do Sistema Nervoso/genética , Autoimunidade/genética , Ciclina A/metabolismo , Fibroblastos/metabolismo , Instabilidade Genômica/genética , Proteínas Monoméricas de Ligação ao GTP/genética , Malformações do Sistema Nervoso/genética , RNA Mensageiro/genética , Doenças Autoimunes do Sistema Nervoso/metabolismo , Proteína Quinase CDC2 , Células Cultivadas , Quinases Ciclina-Dependentes/metabolismo , Dano ao DNA/genética , Dano ao DNA/imunologia , Perfilação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Interferon Tipo I/imunologia , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Malformações do Sistema Nervoso/metabolismo , Fosforilação , Interferência de RNA , Proteína 1 com Domínio SAM e Domínio HD , Transdução de Sinais
4.
Pediatr Rheumatol Online J ; 15(1): 67, 2017 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-28830446

RESUMO

BACKGROUND: Loss-of-function CECR1 mutations cause polyarteritis nodosa (PAN) with childhood onset, an autoinflammatory disorder without significant signs of autoimmunity. Herein we describe the unusual presentation of an autoimmune phenotype with constitutive type I interferon activation in siblings with adenosine deaminase 2 (ADA2) deficiency. CASE PRESENTATION: We describe two siblings with early-onset recurrent strokes, arthritis, oral ulcers, discoid rash, peripheral vascular occlusive disease and high antinuclear antibody titers. Assessment of interferon signatures in blood revealed constitutive type I interferon activation. Aicardi-Goutières syndrome (AGS) was suspected, but no mutation in the known AGS genes were detected. Whole exome sequencing identified compound heterozygosity for a known and a novel mutation in the CECR1 gene. Functional consequences of the mutations were demonstrated by marked reduction in ADA2 catalytic activity. CONCLUSIONS: Our findings demonstrate that ADA2 deficiency can cause an unusual autoimmune phenotype extending the phenotypic spectrum of PAN. Constitutive interferon I activation in patient blood suggests a possible role of type I interferon in disease pathogenesis which may have therapeutic implications.


Assuntos
Adenosina Desaminase/deficiência , Agamaglobulinemia/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Interferon Tipo I/metabolismo , Poliarterite Nodosa/genética , Imunodeficiência Combinada Severa/genética , Adenosina Desaminase/genética , Agamaglobulinemia/complicações , Agamaglobulinemia/diagnóstico , Criança , Pré-Escolar , Humanos , Lactente , Interferon Tipo I/genética , Masculino , Mutação , Linhagem , Fenótipo , Poliarterite Nodosa/complicações , Imunodeficiência Combinada Severa/complicações , Imunodeficiência Combinada Severa/diagnóstico
5.
Nat Commun ; 7: 11752, 2016 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-27230542

RESUMO

Immune recognition of cytosolic DNA represents a central antiviral defence mechanism. Within the host, short single-stranded DNA (ssDNA) continuously arises during the repair of DNA damage induced by endogenous and environmental genotoxic stress. Here we show that short ssDNA traverses the nuclear membrane, but is drawn into the nucleus by binding to the DNA replication and repair factors RPA and Rad51. Knockdown of RPA and Rad51 enhances cytosolic leakage of ssDNA resulting in cGAS-dependent type I IFN activation. Mutations in the exonuclease TREX1 cause type I IFN-dependent autoinflammation and autoimmunity. We demonstrate that TREX1 is anchored within the outer nuclear membrane to ensure immediate degradation of ssDNA leaking into the cytosol. In TREX1-deficient fibroblasts, accumulating ssDNA causes exhaustion of RPA and Rad51 resulting in replication stress and activation of p53 and type I IFN. Thus, the ssDNA-binding capacity of RPA and Rad51 constitutes a cell intrinsic mechanism to protect the cytosol from self DNA.


Assuntos
Citosol/metabolismo , DNA de Cadeia Simples/metabolismo , Rad51 Recombinase/metabolismo , Proteína de Replicação A/metabolismo , Células Cultivadas , DNA de Cadeia Simples/genética , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Células HEK293 , Células HeLa , Humanos , Interferon Tipo I/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ligação Proteica , Interferência de RNA , Rad51 Recombinase/genética , Proteína de Replicação A/genética , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA