Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 206(8): 1691-1696, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33782089

RESUMEN

Severe COVID-19 disease is associated with elevated inflammatory responses. One form of Aicardi-Goutières syndrome caused by inactivating mutations in ADAR results in reduced adenosine-to-inosine (A-to-I) editing of endogenous dsRNAs, induction of IFNs, IFN-stimulated genes, other inflammatory mediators, morbidity, and mortality. Alu elements, ∼10% of the human genome, are the most common A-to-I-editing sites. Using leukocyte whole-genome RNA-sequencing data, we found reduced A-to-I editing of Alu dsRNAs in patients with severe COVID-19 disease. Dendritic cells infected with COVID-19 also exhibit reduced A-to-I editing of Alu dsRNAs. Unedited Alu dsRNAs, but not edited Alu dsRNAs, are potent inducers of IRF and NF-κB transcriptional responses, IL6, IL8, and IFN-stimulated genes. Thus, decreased A-to-I editing that may lead to accumulation of unedited Alu dsRNAs and increased inflammatory responses is associated with severe COVID-19 disease.


Asunto(s)
Adenosina/genética , Elementos Alu/genética , COVID-19/genética , Inosina/genética , Edición de ARN/genética , ARN Bicatenario/genética , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Adenosina/metabolismo , COVID-19/patología , Células Dendríticas/metabolismo , Células Dendríticas/virología , Genoma Humano , Humanos , Inosina/metabolismo , Factores Reguladores del Interferón/metabolismo , FN-kappa B/metabolismo , RNA-Seq , Transducción de Señal/genética
2.
Am J Transplant ; 22(12): 2804-2820, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35997569

RESUMEN

IL-26 is a Th17 cytokine, with its gene being absent in rodents. To characterize the in vivo immunological effects of IL-26 in chronic systemic inflammation, we used human IL26 transgenic (hIL-26Tg) mice and human umbilical cord blood mononuclear cells (hCBMC) in mouse allogeneic-graft-versus-host disease (GVHD) and chronic xenogeneic-GVHD model, respectively. Transfer of bone marrow and spleen T cells from hIL-26Tg mice into B10.BR mice resulted in GVHD progression, with clinical signs of tissue damage in multiple organs. IL-26 markedly increased neutrophil levels both in the GVHD-target tissues and peripheral blood. Expression levels of Th17 cytokines in hIL-26Tg mice-derived donor CD4 T cells were significantly increased, whereas IL-26 did not affect cytotoxic function of donor CD8 T cells. In addition, granulocyte-colony stimulating factor, IL-1ß, and IL-6 levels were particularly enhanced in hIL-26Tg mice. We also developed a humanized neutralizing anti-IL-26 monoclonal antibody (mAb) for therapeutic use, and its administration after onset of chronic xenogeneic-GVHD mitigated weight loss and prolonged survival, with preservation of graft-versus-leukemia effect. Taken together, our data elucidate the in vivo immunological effects of IL-26 in chronic GVHD models and suggest that a humanized anti-IL-26 mAb may be a potential therapeutic agent for the treatment of chronic GVHD.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Ratones , Humanos , Animales , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/etiología , Linfocitos T CD8-positivos , Anticuerpos Monoclonales Humanizados/uso terapéutico , Ratones Transgénicos , Citocinas , Ratones Endogámicos C57BL , Trasplante de Médula Ósea
3.
J Immunol ; 205(10): 2606-2617, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-33046502

RESUMEN

Sensors that detect dsRNA stimulate IFN responses as a defense against viral infection. IFN responses are also well documented in a variety of human autoimmune diseases, including relapsing-remitting multiple sclerosis (MS), in which increased IFN responses result from increased levels of double-stranded endogenous Alu RNAs. Mechanisms underlying increases in double-stranded Alu RNAs in MS are obscure. We find widespread loss of adenosine-to-inosine editing of Alu RNAs in MS. Unedited Alu RNAs are potent activators of both IFN and NF-κB responses via the dsRNA sensors, RIG-I, and TLR3. Minor editing of highly active Alu elements abrogates the ability to activate both transcriptional responses. Thus, adenosine-to-inosine editing may also represent an important defense against autoimmune diseases such as MS.


Asunto(s)
Elementos Alu/inmunología , Esclerosis Múltiple Recurrente-Remitente/genética , Edición de ARN/inmunología , ARN Bicatenario/inmunología , Activación Transcripcional/inmunología , Adenosina/genética , Elementos Alu/genética , Proteína 58 DEAD Box/metabolismo , Conjuntos de Datos como Asunto , Células HEK293 , Humanos , Inflamación/genética , Inflamación/inmunología , Inosina/genética , Interferones/metabolismo , Esclerosis Múltiple Recurrente-Remitente/sangre , Esclerosis Múltiple Recurrente-Remitente/inmunología , FN-kappa B/metabolismo , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , RNA-Seq , Receptores Inmunológicos/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Células THP-1 , Receptor Toll-Like 3/metabolismo , Secuenciación Completa del Genoma
4.
J Autoimmun ; 100: 40-51, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30826177

RESUMEN

Various sensors that detect double-stranded RNA, presumably of viral origin, exist in eukaryotic cells and induce IFN-responses. Ongoing IFN-responses have also been documented in a variety of human autoimmune diseases including relapsing-remitting multiple sclerosis (RRMS) but their origins remain obscure. We find increased IFN-responses in leukocytes in relapsing-remitting multiple sclerosis at distinct stages of disease. Moreover, endogenous RNAs isolated from blood cells of these same patients recapitulate this IFN-response if transfected into naïve cells. These endogenous RNAs are double-stranded RNAs, contain Alu and Line elements and are transcribed from leukocyte transcriptional enhancers. Thus, transcribed endogenous retrotransposon elements can co-opt pattern recognition sensors to induce IFN-responses in RRMS.


Asunto(s)
Elementos Alu/inmunología , Interferones/inmunología , Elementos de Nucleótido Esparcido Largo/inmunología , Esclerosis Múltiple/inmunología , ARN Bicatenario/inmunología , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/patología
5.
Immunity ; 32(5): 581-3, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20510865

RESUMEN

In the periphery, T helper cell differentiation is a key event orchestrating the adaptive immune response yet recent studies demonstrate considerable plasticity in these cell fate decisions. In this issue of Immunity, Mukasa et al. (2010) describe the epigenetic basis underlying this plasticity.

6.
Immunity ; 33(2): 254-65, 2010 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-20691614

RESUMEN

Follicular (FO) and marginal zone (MZ) B cells are maintained in distinct locations within the spleen, but the genetic basis for this separation is still enigmatic. We now report that B cell sequestration requires lineage-specific regulation of migratory receptors by the transcription factor Klf2. Moreover, using gene-targeted mice we show that altered splenic B cell migration confers a significant in vivo gain-of-function phenotype to FO B cells, including the ability to quickly respond to MZ-associated antigens and pathogens in a T cell-dependent manner. This work demonstrates that in wild-type animals, naive FO B cells are actively removed from the MZ, thus restricting their capacity to respond to blood-borne pathogens.


Asunto(s)
Linfocitos B/citología , Linfocitos B/inmunología , Movimiento Celular , Inmunidad Humoral , Bazo/citología , Bazo/inmunología , Animales , Antígenos CD19/genética , Antígenos CD19/inmunología , Antígenos T-Independientes/genética , Antígenos T-Independientes/inmunología , Médula Ósea/inmunología , Diferenciación Celular , Células Cultivadas , Factores de Transcripción de Tipo Kruppel/deficiencia , Factores de Transcripción de Tipo Kruppel/inmunología , Ratones , Ratones Noqueados , Receptores CCR/inmunología
7.
J Immunol ; 199(2): 547-558, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28600289

RESUMEN

We employed whole-genome RNA-sequencing to profile mRNAs and both annotated and novel long noncoding RNAs (lncRNAs) in human naive, central memory, and effector memory CD4+ T cells. Loci transcribing both lineage-specific annotated and novel lncRNA are adjacent to lineage-specific protein-coding genes in the genome. Lineage-specific novel lncRNA loci are transcribed from lineage-specific typical- and supertranscriptional enhancers and are not multiexonic, thus are more similar to enhancer RNAs. Novel enhancer-associated lncRNAs transcribed from the IFNG locus bind the transcription factor NF-κB and enhance binding of NF-κB to the IFNG genomic locus. Depletion of the annotated lncRNA, IFNG-AS1, or one IFNG enhancer-associated lncRNA abrogates IFNG expression by memory T cells, indicating these lncRNAs have biologic function.


Asunto(s)
Memoria Inmunológica , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Linfocitos T/inmunología , Linaje de la Célula , Genoma Humano , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , FN-kappa B/metabolismo , ARN Mensajero/genética , Secuencias Reguladoras de Ácidos Nucleicos , Análisis de Secuencia de ARN
8.
J Immunol ; 197(12): 4509-4517, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27913643

RESUMEN

The central dogma of molecular biology states that DNA makes RNA makes protein. Discoveries over the last quarter of a century found that the process of DNA transcription into RNA gives rise to a diverse array of functional RNA species, including genes that code for protein and noncoding RNAs. For decades, the focus has been on understanding how protein-coding genes are regulated to influence protein expression. However, with the completion of the Human Genome Project and follow-up ENCODE data, it is now appreciated that only 2-3% of the genome codes for protein-coding gene exons and that the bulk of the transcribed genome, apart from ribosomal RNAs, is at the level of noncoding RNA genes. In this article, we focus on the biogenesis and regulation of a distinct class of noncoding RNA molecules termed long, noncoding RNAs in the context of the immune system.


Asunto(s)
Regulación de la Expresión Génica , Sistema Inmunológico , ARN Largo no Codificante , Transcripción Genética , Animales , Ensamble y Desensamble de Cromatina , Genoma , Humanos , ARN Largo no Codificante/biosíntesis , ARN Largo no Codificante/inmunología
9.
J Immunol ; 195(3): 791-5, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26091715

RESUMEN

Of identified genetic variants, HLA polymorphisms confer the greatest risk for developing autoimmune diseases, including rheumatoid arthritis (HLA-DRB1*04). There are strong influences of HLA polymorphisms on cell type-specific gene expression in B cells and monocytes. Their influence on gene expression in CD4(+) T cells is not known. We determined transcript and proteins levels of target genes in lymphocyte/monocyte subsets in healthy controls and rheumatoid arthritis subjects as a function of HLA-DRB1*04 haplotype. We identified gene expression dependent on HLA-DRB1*04 genotype in CD4(+) T cells. NF-κB activity in CD4(+) T cells was also dependent on HLA-DRB1*04 genotype, and blocking HLA-DR inhibited NF-κB activity in CD4(+) T cells and normalized gene expression, as did pharmacologic inhibition of NF-κB. We conclude that interactions between TCR and MHC class II encoded by HLA-DRB1*04 create a proinflammatory "hum" altering CD4(+) T cell phenotype.


Asunto(s)
Artritis Reumatoide/genética , Linfocitos T CD4-Positivos/inmunología , Cadenas HLA-DRB1/genética , Cadenas HLA-DRB1/inmunología , FN-kappa B/metabolismo , Artritis Reumatoide/inmunología , Activación Enzimática , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/metabolismo , Polimorfismo Genético , Receptores de Antígenos de Linfocitos T/inmunología , Factor de Transcripción AP-1/antagonistas & inhibidores , Factor de Transcripción AP-1/metabolismo
10.
J Immunol ; 194(8): 3697-712, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25786689

RESUMEN

Obliterative bronchiolitis is a potentially life-threatening noninfectious pulmonary complication after allogeneic hematopoietic stem cell transplantation and the only pathognomonic manifestation of pulmonary chronic graft-versus-host disease (cGVHD). In the current study, we identified a novel effect of IL-26 on transplant-related obliterative bronchiolitis. Sublethally irradiated NOD/Shi-scidIL2rγ(null) mice transplanted with human umbilical cord blood (HuCB mice) gradually developed clinical signs of graft-versus-host disease (GVHD) such as loss of weight, ruffled fur, and alopecia. Histologically, lung of HuCB mice exhibited obliterative bronchiolitis with increased collagen deposition and predominant infiltration with human IL-26(+)CD26(+)CD4 T cells. Concomitantly, skin manifested fat loss and sclerosis of the reticular dermis in the presence of apoptosis of the basilar keratinocytes, whereas the liver exhibited portal fibrosis and cholestasis. Moreover, although IL-26 is absent from rodents, we showed that IL-26 increased collagen synthesis in fibroblasts and promoted lung fibrosis in a murine GVHD model using IL-26 transgenic mice. In vitro analysis demonstrated a significant increase in IL-26 production by HuCB CD4 T cells following CD26 costimulation, whereas Ig Fc domain fused with the N-terminal of caveolin-1 (Cav-Ig), the ligand for CD26, effectively inhibited production of IL-26. Administration of Cav-Ig before or after onset of GVHD impeded the development of clinical and histologic features of GVHD without interrupting engraftment of donor-derived human cells, with preservation of the graft-versus-leukemia effect. These results therefore provide proof of principle that cGVHD of the lungs is caused in part by IL-26(+)CD26(+)CD4 T cells, and that treatment with Cav-Ig could be beneficial for cGVHD prevention and therapy.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Trasplante de Células Madre de Sangre del Cordón Umbilical , Dipeptidil Peptidasa 4/inmunología , Enfermedad Injerto contra Huésped/inmunología , Interleucinas/inmunología , Enfermedades Pulmonares/inmunología , Pulmón/inmunología , Animales , Linfocitos T CD4-Positivos/patología , Caveolina 1/genética , Caveolina 1/farmacología , Dermis/inmunología , Dermis/patología , Dipeptidil Peptidasa 4/genética , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/patología , Efecto Injerto vs Leucemia/genética , Humanos , Interleucinas/genética , Pulmón/patología , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/patología , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Células 3T3 NIH , Receptores Fc/genética , Receptores Fc/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología
11.
Proc Natl Acad Sci U S A ; 111(26): 9579-84, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24979767

RESUMEN

Regulatory T cells (Tregs) are a specialized subset of CD4(+) T cells that maintain self-tolerance by functionally suppressing autoreactive lymphocytes. The Treg compartment is composed of thymus-derived Tregs (tTregs) and peripheral Tregs (pTregs) that are generated in secondary lymphoid organs after exposure to antigen and specific cytokines, such as TGF-ß. With regard to this latter lineage, pTregs [and their ex vivo generated counterparts, induced Tregs (iTregs)] offer particular therapeutic potential because these cells can be raised against specific antigens to limit autoimmunity. We now report that transcription factor Krüppel-like factor 2 (KLF2) is necessary for the generation of iTregs but not tTregs. Moreover, drugs that limit KLF2 proteolysis during T-cell activation enhance iTreg development. To the authors' knowledge, this study identifies the first transcription factor to distinguish between i/pTreg and tTreg ontogeny and demonstrates that KLF2 is a therapeutic target for the production of regulatory T cells.


Asunto(s)
Autoinmunidad/inmunología , Diferenciación Celular/inmunología , Factores de Transcripción de Tipo Kruppel/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Autoinmunidad/genética , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Citometría de Flujo , Factores de Transcripción de Tipo Kruppel/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
12.
J Immunol ; 193(8): 3959-65, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25225667

RESUMEN

Long noncoding RNAs (lncRNAs), critical regulators of protein-coding genes, are likely to be coexpressed with neighboring protein-coding genes in the genome. How the genome integrates signals to achieve coexpression of lncRNA genes and neighboring protein-coding genes is not well understood. The lncRNA Tmevpg1 (NeST, Ifng-AS1) is critical for Th1-lineage-specific expression of Ifng and is coexpressed with Ifng. In this study, we show that T-bet guides epigenetic remodeling of Tmevpg1 proximal and distal enhancers, leading to recruitment of stimulus-inducible transcription factors, NF-κB and Ets-1, to the locus. Activities of Tmevpg1-specific enhancers and Tmevpg1 transcription are dependent upon NF-κB. Thus, we propose that T-bet stimulates epigenetic remodeling of Tmevpg1-specific enhancers and Ifng-specific enhancers to achieve Th1-lineage-specific expression of Ifng.


Asunto(s)
Elementos de Facilitación Genéticos , Epigénesis Genética , Interferón gamma/genética , ARN Largo no Codificante/genética , Proteínas de Dominio T Box/metabolismo , Células TH1/inmunología , Animales , Linaje de la Célula/inmunología , Sitios Genéticos , Ratones , Ratones Endogámicos BALB C , Proteína Proto-Oncogénica c-ets-1/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Proteínas de Dominio T Box/genética , Factor de Transcripción ReIA/metabolismo
13.
Rheumatology (Oxford) ; 54(1): 178-87, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25118313

RESUMEN

OBJECTIVES: Nuclear factor κB (NF-κB) is a critical activator of inflammatory processes and MTX is one of the most commonly prescribed DMARDs for treatment of RA. We sought to determine whether MTX inhibited NF-κB activity in RA and in lymphocytes and fibroblast-like synoviocytes (FLSs) and to define underlying mechanisms of action. METHODS: An NF-κB luciferase reporter plasmid was used to measure NF-κB activation across experimental stimuli. Flow cytometry was used to quantify changes in intracellular protein levels, measure levels of reactive oxygen species and determine apoptosis. Quantitative RT-PCR was used to identify changes in MTX target genes. RESULTS: In T cell lines, MTX (0.1 µM) inhibited activation of NF-κB via depletion of tetrahydrobiopterin (BH4) and increased Jun-N-terminal kinase (JNK)-dependent p53 activity. Inhibitors of BH4 activity or synthesis also inhibited NF-κB activation and, similar to MTX, increased JNK, p53, p21 and JUN activity. Patients with RA expressed increased levels of phosphorylated or active RelA (p65) compared with controls. Levels of phosphorylated RelA were reduced in patients receiving low-dose MTX therapy. In contrast, inhibition of NF-κB activation by MTX was not mediated via BH4 depletion and JNK activation in FLSs, but rather was completely prevented by adenosine receptor antagonists. CONCLUSION: Our findings support a model whereby distinct pathways are activated by MTX in T cells and FLSs to inhibit NF-κB activation.


Asunto(s)
Fibroblastos/efectos de los fármacos , Metotrexato/farmacología , FN-kappa B/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Membrana Sinovial/efectos de los fármacos , Adulto , Anciano , Apoptosis/efectos de los fármacos , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Estudios de Casos y Controles , Línea Celular , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Células Jurkat , MAP Quinasa Quinasa 4/metabolismo , Masculino , Metotrexato/uso terapéutico , Persona de Mediana Edad , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Membrana Sinovial/metabolismo , Membrana Sinovial/patología
14.
J Immunol ; 188(4): 1726-33, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22246629

RESUMEN

Previous studies have identified multiple conserved noncoding sequences (CNS) at the mouse Ifng locus sufficient for enhancer activity in cell-based assays. These studies do not directly address biology of the human IFNG locus in a genomic setting. IFNG enhancers may be functionally redundant or each may be functionally unique. We test the hypothesis that each IFNG enhancer has a unique necessary function using a bacterial artificial chromosome transgenic model. We find that CNS-30, CNS-4, and CNS+20 are required at distinct stages of Th1 differentiation, whereas CNS-16 has a repressive role in Th1 and Th2 cells. CNS+20 is required for IFN-γ expression by memory Th1 cells and NKT cells. CNS-4 is required for IFN-γ expression by effector Th1 cells. In contrast, CNS-16, CNS-4, and CNS+20 are each partially required for human IFN-γ expression by NK cells. Thus, IFNG CNS enhancers have redundant necessary functions in NK cells but unique necessary functions in Th cells. These results also demonstrate that distinct CNSs are required to transcribe IFNG at each stage of the Th1 differentiation pathway.


Asunto(s)
Interferón gamma/genética , ARN no Traducido/genética , Secuencias Reguladoras de Ácidos Nucleicos , Células TH1/metabolismo , Animales , Diferenciación Celular/genética , Células Cultivadas , Cromosomas Artificiales Bacterianos , Secuencia Conservada , Regulación de la Expresión Génica , Humanos , Interferón gamma/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células T Asesinas Naturales/citología , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Regiones Promotoras Genéticas , Células TH1/citología , Células TH1/inmunología , Células Th2/citología , Células Th2/inmunología , Células Th2/metabolismo
15.
J Immunol ; 189(5): 2084-8, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22851706

RESUMEN

The majority of the genome is noncoding and was thought to be nonfunctional. However, it is now appreciated that transcriptional control of protein coding genes resides within these noncoding regions. Thousands of genes encoding long intergenic noncoding RNAs (lincRNAs) have been recently identified throughout the genome, which positively or negatively regulate transcription of neighboring target genes. Both TMEVPG1 and its mouse ortholog encode lincRNAs and are positioned near the IFN-γ gene (IFNG). In this study, we show that transcription of both mouse and human TMEVPG1 genes is Th1 selective and dependent on Stat4 and T-bet, transcription factors that drive the Th1 differentiation program. Ifng expression is partially restored in Stat4-/-Tbx21-/- cells through coexpression of T-bet and Tmevpg1, and Tmevpg1 expression contributes to, but alone is not sufficient to, drive Th1-dependent Ifng expression. Our results suggest that TMEVPG1 belongs to the general class of lincRNAs that positively regulate gene transcription.


Asunto(s)
Regulación Viral de la Expresión Génica/inmunología , Interferón gamma/genética , ARN no Traducido/inmunología , Células TH1/inmunología , Células TH1/virología , Theilovirus/genética , Theilovirus/inmunología , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Humanos , Interferón gamma/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , ARN Viral/genética , ARN Viral/inmunología , Células TH1/metabolismo , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
16.
Arthritis Rheum ; 64(6): 1780-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22183962

RESUMEN

OBJECTIVE: To assess defects in expression of critical cell cycle checkpoint genes and proteins in patients with rheumatoid arthritis (RA) relative to presence or absence of methotrexate (MTX) treatment, and to investigate the role of JNK in induction of these genes by MTX. METHODS: Flow cytometric analysis was used to quantify changes in levels of intracellular proteins, measure reactive oxygen species (ROS), and determine apoptosis in different lymphoid populations. Quantitative reverse transcription-polymerase chain reaction was used to identify changes in cell cycle checkpoint target genes. RESULTS: RA patients expressed reduced baseline levels of MAPK9, TP53, CDKN1A, CDKN1B, CHEK2, and RANGAP1 messenger RNA (mRNA) and JNK total protein. The reduction in expression of mRNA for MAPK9, TP53, CDKN1A, and CDKN1B was greater in patients not receiving MTX than in those receiving low-dose MTX, with no difference in expression levels of CHEK2 and RANGAP1 mRNA between MTX-treated and non-MTX-treated patients. Further, JNK levels were inversely correlated with C-reactive protein levels in RA patients. In tissue culture, MTX induced expression of both p53 and p21 by JNK-2- and JNK-1-dependent mechanisms, respectively, while CHEK2 and RANGAP1 were not induced by MTX. MTX also induced ROS production, JNK activation, and sensitivity to apoptosis in activated T cells. Supplementation with tetrahydrobiopterin blocked these MTX-mediated effects. CONCLUSION: Our findings support the notion that MTX restores some, but not all, of the proteins contributing to cell cycle checkpoint deficiencies in RA T cells, via a JNK-dependent pathway.


Asunto(s)
Antirreumáticos/farmacología , Artritis Reumatoide/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metotrexato/farmacología , Adulto , Anciano , Antirreumáticos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/genética , Puntos de Control del Ciclo Celular/fisiología , Femenino , Citometría de Flujo , Humanos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Metotrexato/uso terapéutico , Persona de Mediana Edad , Especies Reactivas de Oxígeno/metabolismo
17.
Cancer Res Commun ; 3(9): 1800-1809, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37691856

RESUMEN

It was recently found that patients with relapsing remitting multiple sclerosis exhibit widespread loss of adenosine-to-inosine (A-to-I) RNA editing, which contributes to the accumulation of immunostimulatory double-stranded Alu RNA in circulating leukocytes and an attendant increase in levels of proinflammatory cytokines (e.g., type I IFNs). A specific Alu RNA (i.e., AluJb RNA) was implicated in activating multiple RNA-sensing pathways and found to be a potent innate immune agonist. Here, we have performed a bioinformatic analysis of A-to-I RNA editing in human melanoma samples and determined that pre-therapy levels of A-to-I RNA editing negatively correlate with survival times, suggesting that an accumulation of endogenous double-stranded Alu RNA might contribute to cancer patient survival. Furthermore, we demonstrated that immunostimulatory Alu RNA can be leveraged pharmacologically for cancer immunotherapy. AluJb RNA was in vitro transcribed and then formulated with endosome-destabilizing polymer nanoparticles to improve intracellular delivery of the RNA and enable activation of RNA-sensing pathways. AluJb RNA/polymer complexes (i.e., Alu-NPs) were engineered to form colloidally stable nanoparticles that exhibited immunostimulatory activity in vitro and in vivo. Finally, the therapeutic potential of Alu-NPs for the treatment of cancer was demonstrated by attenuated tumor growth and prolonged survival in the B16.F10 murine melanoma tumor model. Thus, these data collectively implicate intratumoral Alu RNA as a potentiator of antitumor innate immunity and identify AluJb RNA as a novel nucleic acid immunotherapeutic for cancer. Significance: Loss of A-to-I editing leads to accumulation of unedited Alu RNAs that activate innate immunity via RNA-sensing pattern recognition receptors. When packaged into endosome-releasing polymer nanoparticles, AluJB RNA becomes highly immunostimulatory and can be used pharmacologically to inhibit tumor growth in mouse melanoma models. These findings identify Alu RNAs as a new class of nucleic acid innate immune agonists for cancer immunotherapy.


Asunto(s)
Melanoma , Ácidos Nucleicos , Humanos , Animales , Ratones , Inmunoterapia , Inmunización , ARN Bicatenario , Melanoma/genética
18.
Bioinformatics ; 27(5): 686-92, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21266443

RESUMEN

MOTIVATION: In genome-wide association studies (GWAS) of complex diseases, genetic variants having real but weak associations often fail to be detected at the stringent genome-wide significance level. Pathway analysis, which tests disease association with combined association signals from a group of variants in the same pathway, has become increasingly popular. However, because of the complexities in genetic data and the large sample sizes in typical GWAS, pathway analysis remains to be challenging. We propose a new statistical model for pathway analysis of GWAS. This model includes a fixed effects component that models mean disease association for a group of genes, and a random effects component that models how each gene's association with disease varies about the gene group mean, thus belongs to the class of mixed effects models. RESULTS: The proposed model is computationally efficient and uses only summary statistics. In addition, it corrects for the presence of overlapping genes and linkage disequilibrium (LD). Via simulated and real GWAS data, we showed our model improved power over currently available pathway analysis methods while preserving type I error rate. Furthermore, using the WTCCC Type 1 Diabetes (T1D) dataset, we demonstrated mixed model analysis identified meaningful biological processes that agreed well with previous reports on T1D. Therefore, the proposed methodology provides an efficient statistical modeling framework for systems analysis of GWAS. AVAILABILITY: The software code for mixed models analysis is freely available at http://biostat.mc.vanderbilt.edu/LilyWang.


Asunto(s)
Estudio de Asociación del Genoma Completo/métodos , Modelos Lineales , Programas Informáticos , Simulación por Computador , Genotipo , Humanos , Desequilibrio de Ligamiento , Polimorfismo de Nucleótido Simple
19.
Arthritis Rheum ; 63(9): 2606-16, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21618198

RESUMEN

OBJECTIVE: Low-dose methotrexate (MTX) is an effective therapy for rheumatoid arthritis (RA), yet its mechanism of action is incompletely understood. The aim of this study was to explore the induction of apoptosis by MTX. METHODS: Flow cytometry was performed to assess changes in the levels of intracellular proteins, reactive oxygen species (ROS), and apoptosis. Quantitative polymerase chain reaction was performed to assess changes in the transcript levels of select target genes in response to MTX. RESULTS: MTX did not directly induce apoptosis but rather "primed" cells for markedly increased sensitivity to apoptosis via either mitochondrial or death receptor pathways, by a JNK-dependent mechanism. Increased sensitivity to apoptosis was mediated, at least in part, by MTX-dependent production of ROS, JNK activation, and JNK-dependent induction of genes whose protein products promote apoptosis. Supplementation with tetrahydrobiopterin blocked these MTX-induced effects. Patients with RA who were receiving low-dose MTX therapy expressed elevated levels of the JNK target gene, jun. CONCLUSION: Our results support a model whereby MTX inhibits reduction of dihydrobiopterin to tetrahydrobiopterin, resulting in increased production of ROS, increased JNK activity, and increased sensitivity to apoptosis. The finding of increased jun levels in patients with RA receiving low-dose MTX supports the notion that this pathway is activated by MTX in vivo and may contribute to the efficacy of MTX in inflammatory disease.


Asunto(s)
Antirreumáticos/uso terapéutico , Apoptosis/efectos de los fármacos , Artritis Reumatoide/tratamiento farmacológico , Expresión Génica/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Metotrexato/uso terapéutico , Adulto , Antirreumáticos/farmacología , Apoptosis/fisiología , Artritis Reumatoide/metabolismo , Biopterinas/análogos & derivados , Biopterinas/farmacología , Línea Celular Tumoral , Humanos , Metotrexato/farmacología , Persona de Mediana Edad , Especies Reactivas de Oxígeno/metabolismo , Células Tumorales Cultivadas
20.
J Immunol ; 185(3): 1492-501, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20574006

RESUMEN

Genes, such as IFNG, which are expressed in multiple cell lineages of the immune system, may employ a common set of regulatory elements to direct transcription in multiple cell types or individual regulatory elements to direct expression in individual cell lineages. By employing a bacterial artificial chromosome transgenic system, we demonstrate that IFNG employs unique regulatory elements to achieve lineage-specific transcriptional control. Specifically, a one 1-kb element 30 kb upstream of IFNG activates transcription in T cells and NKT cells but not in NK cells. This distal regulatory element is a Runx3 binding site in Th1 cells and is needed for RNA polymerase II recruitment to IFNG, but it is not absolutely required for histone acetylation of the IFNG locus. These results support a model whereby IFNG uses cis-regulatory elements with cell type-restricted function.


Asunto(s)
Linaje de la Célula/genética , Linaje de la Célula/inmunología , Regulación de la Expresión Génica/inmunología , Sitios Genéticos/inmunología , Interferón gamma/biosíntesis , Interferón gamma/genética , Animales , Células Cultivadas , Secuencia Conservada/genética , Secuencia Conservada/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Células Asesinas Naturales/enzimología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células T Asesinas Naturales/enzimología , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Transporte de Proteínas/genética , Transporte de Proteínas/inmunología , ARN Polimerasa II/metabolismo , Elementos Reguladores de la Transcripción/inmunología , Células TH1/enzimología , Células TH1/inmunología , Células TH1/metabolismo , Sitio de Iniciación de la Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA