Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Cell Immunol ; 357: 104201, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32979763

RESUMEN

The DNA binding protein AT-rich interacting domain 3a (ARID3a)2 is expressed in healthy human hematopoietic cord blood progenitors where its modulation influences myeloid versus B lineage development. ARID3a is also variably expressed in subsets of adult peripheral blood hematopoietic progenitors where the consequences of ARID3a expression are unknown. In B lymphocytes, Toll-like receptor (TLR)3 signaling induces ARID3a expression in association with Type I interferon inflammatory cytokines. We hypothesized that TLR ligand stimulation of peripheral blood hematopoietic progenitors would induce ARID3a expression resulting in interferon production, and potentially influencing lineage decisions. Our data revealed that the TLR9 agonist CpG induces ARID3a expression with interferon alpha synthesis in human hematopoietic progenitors. However, ARID3a expression was not associated with increased B lineage development. These results demonstrate the need for further experiments to better define how pathogen-associated responses influence hematopoiesis.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Células Madre Hematopoyéticas/metabolismo , Interferón-alfa/biosíntesis , Receptores Toll-Like/sangre , Factores de Transcripción/biosíntesis , Adulto , Linfocitos B/citología , Linfocitos B/metabolismo , Islas de CpG , Citocinas/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/sangre , Femenino , Citometría de Flujo/métodos , Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Interferón-alfa/genética , Interferón-alfa/inmunología , Ligandos , Persona de Mediana Edad , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/sangre
2.
Immun Ageing ; 17: 24, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32905435

RESUMEN

BACKGROUND: Immunologic aging leads to immune dysfunction, significantly reducing the quality of life of the elderly. Aged-related defects in early hematopoiesis result in reduced lymphoid cell development, functionally defective mature immune cells, and poor protective responses to vaccines and pathogens. Despite considerable progress understanding the underlying causes of decreased immunity in the elderly, the mechanisms by which these occur are still poorly understood. The DNA-binding protein ARID3a is expressed in a subset of human hematopoietic progenitors. Inhibition of ARID3a in bulk human cord blood CD34+ hematopoietic progenitors led to developmental skewing toward myeloid lineage at the expense of lymphoid lineage cells in vitro. Effects of ARID3a expression in adult-derived hematopoietic stem cells (HSCs) have not been analyzed, nor has ARID3a expression been assessed in relationship to age. We hypothesized that decreases in ARID3a could explain some of the defects observed in aging. RESULTS: Our data reveal decreased frequencies of ARID3a-expressing peripheral blood HSCs from aged healthy individuals compared with young donor HSCs. Inhibition of ARID3a in young donor-derived HSCs limits B lineage potential, suggesting a role for ARID3a in B lymphopoiesis in bone marrow-derived HSCs. Increasing ARID3a levels of HSCs from aged donors in vitro alters B lineage development and maturation. Finally, single cell analyses of ARID3a-expressing HSCs from young versus aged donors identify a number of differentially expressed genes in aged ARID3A-expressing cells versus young ARID3A-expressing HSCs, as well as between ARID3A-expressing and non-expressing cells in both young and aged donor HSCs. CONCLUSIONS: These data suggest that ARID3a-expressing HSCs from aged individuals differ at both molecular and functional levels compared to ARID3a-expressing HSCs from young individuals.

3.
J Autoimmun ; 96: 158-167, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30297159

RESUMEN

Type I interferons (IFN) causes inflammatory responses to pathogens, and can be elevated in autoimmune diseases such as systemic lupus erythematosus (SLE). We previously reported unexpected associations of increased numbers of B lymphocytes expressing the DNA-binding protein ARID3a with both IFN alpha (IFNα) expression and increased disease activity in SLE. Here, we determined that IFNα producing low density neutrophils (LDNs) and plasmacytoid dendritic cells (pDCs) from SLE patients exhibit strong associations between ARID3a protein expression and IFNα production. Moreover, SLE disease activity indices correlate most strongly with percentages of ARID3a+ LDNs, but were also associated, less significantly, with IFNα expression in LDNs and pDCs. Hierarchical clustering and transcriptome analyses of LDNs and pDCs revealed SLE patients with low ARID3a expression cluster with healthy controls and identified gene profiles associated with increased proportions of ARID3a- and IFNα-expressing cells of each type. These data identify ARID3a as a potential transcription regulator of IFNα-related inflammatory responses and other pathways important for SLE disease activity.


Asunto(s)
Linfocitos B/fisiología , Proteínas de Unión al ADN/genética , Células Dendríticas/fisiología , Interferón-alfa/metabolismo , Lupus Eritematoso Sistémico/genética , Neutrófilos/fisiología , Factores de Transcripción/genética , Adulto , Anciano , Progresión de la Enfermedad , Femenino , Regulación de la Expresión Génica , Estudios de Asociación Genética , Humanos , Inmunidad Innata , Interferón-alfa/genética , Lupus Eritematoso Sistémico/inmunología , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Transcriptoma
4.
J Immunol ; 196(2): 614-23, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26685208

RESUMEN

We recently reported that the transcription factor ARID3a is expressed in a subset of human hematopoietic progenitor stem cells in both healthy individuals and in patients with systemic lupus erythematosus. Numbers of ARID3a(+) lupus hematopoietic stem progenitor cells were associated with increased production of autoreactive Abs when those cells were introduced into humanized mouse models. Although ARID3a/Bright knockout mice died in utero, they exhibited decreased numbers of hematopoietic stem cells and erythrocytes, indicating that ARID3a is functionally important for hematopoiesis in mice. To explore the requirement for ARID3a for normal human hematopoiesis, hematopoietic stem cell progenitors from human cord blood were subjected to both inhibition and overexpression of ARID3a in vitro. Inhibition of ARID3a resulted in decreased B lineage cell production accompanied by increases in cells with myeloid lineage markers. Overexpression of ARID3a inhibited both myeloid and erythroid differentiation. Additionally, inhibition of ARID3a in hematopoietic stem cells resulted in altered expression of transcription factors associated with hematopoietic lineage decisions. These results suggest that appropriate regulation of ARID3a is critical for normal development of both myeloid and B lineage pathways.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Factores de Transcripción/metabolismo , Linaje de la Célula , Sangre Fetal , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Humanos , Técnicas In Vitro , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Transducción Genética
5.
Am J Hum Genet ; 94(4): 586-98, 2014 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-24702955

RESUMEN

Efforts to identify lupus-associated causal variants in the FAM167A/BLK locus on 8p21 are hampered by highly associated noncausal variants. In this report, we used a trans-population mapping and sequencing strategy to identify a common variant (rs922483) in the proximal BLK promoter and a tri-allelic variant (rs1382568) in the upstream alternative BLK promoter as putative causal variants for association with systemic lupus erythematosus. The risk allele (T) at rs922483 reduced proximal promoter activity and modulated alternative promoter usage. Allelic differences at rs1382568 resulted in altered promoter activity in B progenitor cell lines. Thus, our results demonstrated that both lupus-associated functional variants contribute to the autoimmune disease association by modulating transcription of BLK in B cells and thus potentially altering immune responses.


Asunto(s)
Lupus Eritematoso Sistémico/genética , Regiones Promotoras Genéticas , Transcripción Genética , Familia-src Quinasas/genética , Alelos , Cromosomas Humanos Par 8 , Ensayo de Cambio de Movilidad Electroforética , Femenino , Predisposición Genética a la Enfermedad , Haplotipos , Humanos , Masculino , Polimorfismo de Nucleótido Simple
6.
J Immunol ; 194(3): 940-9, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25535283

RESUMEN

Although hematopoietic stem/progenitor cells (HSPCs) are used for transplantation, characterization of the multiple subsets within this population in humans has lagged behind similar studies in mice. We found that expression of the DNA-binding protein, ARID3a, in mouse stem cells was important for normal development of hematopoietic lineages; however, progenitors expressing ARID3a in humans have not been defined. We previously showed increased numbers of ARID3a(+) B cells in nearly half of systemic lupus erythematosus (SLE) patients, and total numbers of ARID3a(+) B cells were associated with increased disease severity. Because expression of ARID3a in those SLE patients occurred throughout all B cell subsets, we hypothesized that ARID3a expression in patient HSPCs might also be increased relative to expression in healthy controls. Our data now show that ARID3a expression is not limited to any defined subset of HSPCs in either healthy controls or SLE patients. Numbers of ARID3a(+) HSPCs in SLE patients were increased over numbers of ARID3a(+) cells in healthy controls. Although all SLE-derived HSPCs exhibited poor colony formation in vitro compared with controls, SLE HSPCs with high numbers of ARID3a(+) cells yielded increased numbers of cells expressing the early progenitor marker, CD34. SLE HSPCs with high numbers of ARID3a(+) cells also more readily generated autoantibody-producing cells than HSPCs with lower levels of ARID3a in a humanized mouse model. These data reveal new functions for ARID3a in early hematopoiesis and suggest that knowledge regarding ARID3a levels in HSPCs could be informative for applications requiring transplantation of those cells.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Factores de Transcripción/genética , Adulto , Anciano , Animales , Anticuerpos Antinucleares/inmunología , Formación de Anticuerpos/genética , Formación de Anticuerpos/inmunología , Antígenos de Superficie/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Estudios de Casos y Controles , Expresión Génica , Humanos , Inmunofenotipificación , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Persona de Mediana Edad , Adulto Joven
7.
J Autoimmun ; 75: 130-140, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27522115

RESUMEN

Previously, we determined that enhanced disease activity in patients with systemic lupus erythematosus (SLE) was associated with dramatic increases in numbers of B lymphocytes expressing the transcription factor ARID3a. Our data now indicate ARID3a is important for interferon alpha (IFNa) expression and show a strong association between ARID3a expression and transcription of genes associated with lupus IFN signatures. Furthermore, both ARID3a and IFNa production were elicited in healthy control B cells upon stimulation with the TLR 9 agonist, CpG. Importantly, secretion of IFNa from ARID3a+ healthy B lymphocytes stimulated increased IFNa production in plasmacytoid dendritic cells. These data identify ARID3a+ B cells as a novel type of effector B cell, and link ARID3a expression in B lymphocytes to IFN-associated inflammatory responses in SLE.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Proteínas de Unión al ADN/inmunología , Expresión Génica/inmunología , Interferón-alfa/inmunología , Factores de Transcripción/inmunología , Subgrupos de Linfocitos B/efectos de los fármacos , Subgrupos de Linfocitos B/metabolismo , Línea Celular Tumoral , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Humanos , Interferón-alfa/sangre , Interferón-alfa/metabolismo , Oligodesoxirribonucleótidos/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/inmunología , Receptor Toll-Like 9/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Biochem Biophys Res Commun ; 463(4): 1334-1340, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26111446

RESUMEN

Despite exciting new possibilities for regenerative therapy posed by the ability to induce pluripotent stem cells, recapitulation of three-dimensional kidneys for repair or replacement has not been possible. ARID3a-deficient mouse tissues generated multipotent, developmentally plastic cells. Therefore, we assessed the adult mouse ARID3a-/- kidney cell line, KKPS5, which expresses renal progenitor surface markers as an alternative cell source for modeling kidney development. Remarkably, these cells spontaneously developed into multicellular nephron-like structures in vitro, and engrafted into immunocompromised medaka mesonephros, where they formed mouse nephron structures. These data implicate KKPS5 cells as a new model system for studying kidney development.


Asunto(s)
Riñón/anatomía & histología , Animales , División Celular , Línea Celular , Proteínas de Unión al ADN/genética , Riñón/citología , Ratones , Ratones Noqueados , Oryzias , Células Madre/citología , Factores de Transcripción/genética
9.
EMBO J ; 28(6): 711-24, 2009 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-19214191

RESUMEN

Regulation of BCR signalling strength is crucial for B-cell development and function. Bright is a B-cell-restricted factor that complexes with Bruton's tyrosine kinase (Btk) and its substrate, transcription initiation factor-I (TFII-I), to activate immunoglobulin heavy chain gene transcription in the nucleus. Here we show that a palmitoylated pool of Bright is diverted to lipid rafts of resting B cells where it associates with signalosome components. After BCR ligation, Bright transiently interacts with sumoylation enzymes, blocks calcium flux and phosphorylation of Btk and TFII-I and is then discharged from lipid rafts as a Sumo-I-modified form. The resulting lipid raft concentration of Bright contributes to the signalling threshold of B cells, as their sensitivity to BCR stimulation decreases as the levels of Bright increase. Bright regulates signalling independent of its role in IgH transcription, as shown by specific dominant-negative titration of rafts-specific forms. This study identifies a BCR tuning mechanism in lipid rafts that is regulated by differential post-translational modification of a transcription factor with implications for B-cell tolerance and autoimmunity.


Asunto(s)
Microdominios de Membrana/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Agammaglobulinemia Tirosina Quinasa , Animales , Antígenos/metabolismo , Linfocitos B/enzimología , Proteínas de Unión al ADN , Humanos , Inmunoglobulina M/genética , Inmunoglobulina M/metabolismo , Lipoilación , Activación de Linfocitos , Microdominios de Membrana/enzimología , Ratones , Mutación/genética , Oncogenes , Fosforilación , Unión Proteica , Transporte de Proteínas , Proteínas Tirosina Quinasas/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Factores de Transcripción TFII/metabolismo , Transcripción Genética
10.
Cell Immunol ; 272(2): 175-81, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22082566

RESUMEN

Lethal toxin, a key virulence factor produced by Bacillus anthracis, induces cell death, in part by disrupting numerous signaling pathways, in mouse macrophages. However, exposure to sublethal doses of lethal toxin allows some cells to survive. Because these pro-survival signaling events occur within a few hours after exposure to sublethal doses, we hypothesized that acute phase proteins might influence macrophage survival. Our data show that serum amyloid A (SAA) is produced in response to lethal toxin treatment. Moreover, pre-treatment of macrophages with exogenous SAA protected macrophages from lethal toxin-mediated death. Exogenous SAA activated the p38 mitogen activated protein kinase (MAP) kinase pathway, while lethal toxin mutants incapable of p38 activation were incapable of causing cell death. Chemical inhibition of the p38 activation pathway abrogated the protective effects of SAA. These data show that SAA affords protection against lethal toxin in mouse macrophages and link this response to the p38 pathway.


Asunto(s)
Toxinas Bacterianas/farmacología , Macrófagos/efectos de los fármacos , Proteína Amiloide A Sérica/metabolismo , Animales , Antígenos Bacterianos/metabolismo , Bacillus anthracis/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/inmunología , Supervivencia Celular/fisiología , Células Cultivadas , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mutación/genética , Sustancias Protectoras/metabolismo , Proteínas Recombinantes/metabolismo , Proteína Amiloide A Sérica/farmacología , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Stem Cells ; 28(9): 1560-7, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20680960

RESUMEN

B-cell regulator of immunoglobulin heavy chain transcription (Bright)/ARID3a, an A+T-rich interaction domain protein, was originally discovered in B lymphocyte lineage cells. However, expression patterns and high lethality levels in knockout mice suggested that it had additional functions. Three independent lines of evidence show that functional inhibition of Bright results in increased developmental plasticity. Bright-deficient cells from two mouse models expressed a number of pluripotency-associated gene products, expanded indefinitely, and spontaneously differentiated into cells of multiple lineages. Furthermore, direct knockdown of human Bright resulted in colonies capable of expressing multiple lineage markers. These data suggest that repression of this single molecule confers adult somatic cells with new developmental options.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Proteínas de Unión al ADN/deficiencia , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/deficiencia , Animales , Biomarcadores/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes Dominantes , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Interferencia de ARN , Teratoma/genética , Teratoma/metabolismo , Factores de Transcripción/genética
12.
Immunohorizons ; 5(10): 802-817, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34663594

RESUMEN

ARID3a is a DNA-binding protein important for normal hematopoiesis in mice and for in vitro lymphocyte development in human cultures. ARID3a knockout mice die in utero with defects in both early hematopoietic stem cell populations and erythropoiesis. Recent transcriptome analyses in human erythropoietic systems revealed increases in ARID3a transcripts implicating potential roles for ARID3a in human erythrocyte development. However, ARID3a transcript levels do not faithfully reflect protein levels in many cells, and the functions and requirements for ARID3a protein in those systems have not been explored. We used the erythroleukemic cell line K562 as a model to elucidate functions of ARID3a protein in early human erythropoiesis. ARID3a knockdown of hemin-stimulated K562 cells resulted in lack of fetal globin production and modifications in gene expression. Temporal RNA sequencing data link ARID3a expression with the important erythroid regulators Gata1, Gata2, and Klf1 Ablation of ARID3a using CRISPR-Cas9 further demonstrated it is required to maintain chromatin structures associated with erythropoietic differentiation potential. These data demonstrate that the ARID3a protein is required for early erythropoietic events and provide evidence for the requirement of ARID3a functions for proper maintenance of appropriate chromatin structures.


Asunto(s)
Cromatina/metabolismo , Proteínas de Unión al ADN/deficiencia , Eritropoyesis/genética , Factores de Transcripción/deficiencia , Sistemas CRISPR-Cas/genética , Cromatina/genética , Secuenciación de Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Globulinas Fetales/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Células K562 , RNA-Seq , Factores de Transcripción/genética
13.
J Immunol ; 181(10): 6913-22, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18981111

RESUMEN

The transcription factor Bright up-regulates Ig H chain production from select V region promoters and requires Bright dimerization, Bruton's tyrosine kinase (Btk), and the Btk substrate, TFII-I, for this activity. Defects in Btk cause X-linked immunodeficiency disease in mice and humans. Btk-deficient mice exhibit decreased serum IgM production, B cell developmental blocks, absence of peritoneal B1 cells, and subnormal immune responses against Ags, including phosphorylcholine, which confer protection against Streptococcus pneumoniae. Transgenic mice expressing dominant-negative Bright share similarities with Btk-deficient mice, including decreased serum IgM, poor anti-phosphorylcholine responses, and slightly reduced numbers of mature B cells. Although dominant-negative Bright mice developed B1 B cells, these were functionally deficient in Ig secretion. These data suggest a mechanistic explanation for the abnormal responses to phosphorylcholine observed in Btk-deficient mice, and indicate that Bright functions in a subset of Btk-dependent pathways in vivo, particularly those responses dominated by B1 B cells.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Linfocitos B/inmunología , Oncogenes/genética , Transactivadores/genética , Transactivadores/metabolismo , Agammaglobulinemia Tirosina Quinasa , Secuencia de Aminoácidos , Animales , Anticuerpos/sangre , Antígenos CD19/genética , Western Blotting , Proteínas de Unión al ADN , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Proteínas Tirosina Quinasas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Factores de Transcripción
14.
Mol Cell Biol ; 26(12): 4758-68, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16738337

RESUMEN

Bright/ARID3a/Dril1, a member of the ARID family of transcription factors, is expressed in a highly regulated fashion in B lymphocytes, where it enhances immunoglobulin transcription three- to sixfold. Recent publications from our lab indicated that functional, but not kinase-inactive, Bruton's tyrosine kinase (Btk) is critical for Bright activity in an in vitro model system, yet Bright itself is not appreciably tyrosine phosphorylated. These data suggested that a third protein, and Btk substrate, must contribute to Bright-enhanced immunoglobulin transcription. The ubiquitously expressed transcription factor TFII-I was identified as a substrate for Btk several years ago. In this work, we show that TFII-I directly interacts with human Bright through amino acids in Bright's protein interaction domain and that specific tyrosine residues of TFII-I are essential for Bright-induced activity of an immunoglobulin reporter gene. Moreover, inhibition of TFII-I function in a B-cell line resulted in decreased heavy-chain transcript levels. These data suggest that Bright functions as a three-component protein complex in the immunoglobulin locus and tie together previous data indicating important roles for Btk and TFII-I in B lymphocytes.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Genes de Inmunoglobulinas , Cadenas Pesadas de Inmunoglobulina/genética , Transactivadores/metabolismo , Factores de Transcripción TFII/metabolismo , Agammaglobulinemia Tirosina Quinasa , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Secuencia de Bases , Sitios de Unión , Línea Celular , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Humanos , Ratones , Mutación , Oncogenes/genética , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transactivadores/química , Transactivadores/genética , Factores de Transcripción , Factores de Transcripción TFII/genética , Transcripción Genética
15.
Cells ; 8(10)2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31554207

RESUMEN

Systemic lupus erythematosus (SLE) is a devastating and heterogeneous autoimmune disease that affects multiple organs, and for which the underlying causes are unknown. The majority of SLE patients produce autoantibodies, have increased levels of type-I inflammatory cytokines, and can develop glomerulonephritis. Recent studies indicate an unexpected but strong association between increased disease activity in SLE patients and the expression of the DNA-binding protein ARID3a (A + T rich interaction domain protein 3a) in a number of peripheral blood cell types. ARID3a expression was first associated with autoantibody production in B cells; however, more recent findings also indicate associations with expression of the inflammatory cytokine interferon alpha in SLE plasmacytoid dendritic cells and low-density neutrophils. In addition, ARID3a is expressed in hematopoietic stem cells and some adult kidney progenitor cells. SLE cells expressing enhanced ARID3a levels show differential gene expression patterns compared with homologous healthy control cells, identifying new pathways potentially regulated by ARID3a. The associations of ARID3a expression with increased disease severity in SLE, suggest that it, or its downstream targets, may provide new therapeutic targets for SLE.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/terapia , Terapia Molecular Dirigida/tendencias , Factores de Transcripción/fisiología , Adulto , Células Madre Adultas/fisiología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Proteínas de Unión al ADN/genética , Células Dendríticas/fisiología , Humanos , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Terapia Molecular Dirigida/métodos , Factores de Transcripción/genética
16.
Front Immunol ; 10: 1066, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31164884

RESUMEN

Systemic lupus erythematosus (SLE) is an autoimmune disease with a strong genetic component. We recently identified a novel SLE susceptibility locus near RASGRP1, which governs the ERK/MAPK kinase cascade and B-/T-cell differentiation and development. However, precise causal RASGRP1 functional variant(s) and their mechanisms of action in SLE pathogenesis remain undefined. Our goal was to fine-map this locus, prioritize genetic variants likely to be functional, experimentally validate their biochemical mechanisms, and determine the contribution of these SNPs to SLE risk. We performed a meta-analysis across six Asian and European cohorts (9,529 cases; 22,462 controls), followed by in silico bioinformatic and epigenetic analyses to prioritize potentially functional SNPs. We experimentally validated the functional significance and mechanism of action of three SNPs in cultured T-cells. Meta-analysis identified 18 genome-wide significant (p < 5 × 10-8) SNPs, mostly concentrated in two haplotype blocks, one intronic and the other intergenic. Epigenetic fine-mapping, allelic, eQTL, and imbalance analyses predicted three transcriptional regulatory regions with four SNPs (rs7170151, rs11631591-rs7173565, and rs9920715) prioritized for functional validation. Luciferase reporter assays indicated significant allele-specific enhancer activity for intronic rs7170151 and rs11631591-rs7173565 in T-lymphoid (Jurkat) cells, but not in HEK293 cells. Following up with EMSA, mass spectrometry, and ChIP-qPCR, we detected allele-dependent interactions between heterogeneous nuclear ribonucleoprotein K (hnRNP-K) and rs11631591. Furthermore, inhibition of hnRNP-K in Jurkat and primary T-cells downregulated RASGRP1 and ERK/MAPK signaling. Comprehensive association, bioinformatics, and epigenetic analyses yielded putative functional variants of RASGRP1, which were experimentally validated. Notably, intronic variant (rs11631591) is located in a cell type-specific enhancer sequence, where its risk allele binds to the hnRNP-K protein and modulates RASGRP1 expression in Jurkat and primary T-cells. As risk allele dosage of rs11631591 correlates with increased RASGRP1 expression and ERK activity, we suggest that this SNP may underlie SLE risk at this locus.


Asunto(s)
Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos/fisiología , Predisposición Genética a la Enfermedad , Factores de Intercambio de Guanina Nucleótido/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo K/fisiología , Lupus Eritematoso Sistémico/genética , Polimorfismo de Nucleótido Simple , Proteínas de Unión al ADN/análisis , Humanos , Lupus Eritematoso Sistémico/etiología , Sitios de Carácter Cuantitativo
17.
Mol Cell Biol ; 25(6): 2073-84, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15743806

RESUMEN

Bright (B-cell regulator of immunoglobulin heavy chain transcription) binding to immunoglobulin heavy chain loci after B-cell activation is associated with increased heavy chain transcription. Our earlier reports demonstrated that Bright coimmunoprecipitates with Bruton's tyrosine kinase (Btk) and that these proteins associate in a DNA-binding complex in primary B cells. B cells from immunodeficient mice with a mutation in Btk failed to produce stable Bright DNA-binding complexes. In order to determine if Btk is important for Bright function, a transcription activation assay was established and analyzed using real-time PCR technology. Cells lacking both Bright and Btk were transfected with Bright and/or Btk along with an immunoglobulin heavy chain reporter construct. Immunoglobulin gene transcription was enhanced when Bright and Btk were coexpressed. In contrast, neither Bright nor Btk alone led to activation of heavy chain transcription. Furthermore, Bright function required both Btk kinase activity and sequences within the pleckstrin homology domain of Btk. Bright was not appreciably phosphorylated by Btk; however, a third tyrosine-phosphorylated protein coprecipitated with Bright. Thus, the ability of Bright to enhance immunoglobulin transcription critically requires functional Btk.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Cadenas Pesadas de Inmunoglobulina/genética , Oncogenes/fisiología , Proteínas Tirosina Quinasas/fisiología , Transactivadores/fisiología , Activación Transcripcional/fisiología , Agammaglobulinemia Tirosina Quinasa , Secuencias de Aminoácidos/genética , Animales , Proteínas Sanguíneas/genética , Células CHO , Cricetinae , Cricetulus , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Genes Reporteros/genética , Inmunoprecipitación , Linfocitos/metabolismo , Ratones , Oncogenes/genética , Fosfoproteínas/genética , Fosforilación , Regiones Promotoras Genéticas/genética , Estructura Terciaria de Proteína/genética , Proteínas Tirosina Quinasas/genética , Eliminación de Secuencia/genética , Transactivadores/genética , Factores de Transcripción , Transcripción Genética/genética , Activación Transcripcional/genética , Transfección
18.
Front Immunol ; 9: 3064, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30697210

RESUMEN

Memory B cells that are generated during an infection or following vaccination act as sentinels to guard against future infections. Upon repeat antigen exposure memory B cells differentiate into new antibody-secreting plasma cells to provide rapid and sustained protection. Some pathogens evade or suppress the humoral immune system, or induce memory B cells with a diminished ability to differentiate into new plasma cells. This leaves the host vulnerable to chronic or recurrent infections. Single cell approaches coupled with next generation antibody gene sequencing facilitate a detailed analysis of the pathogen-specific memory B cell repertoire. Monoclonal antibodies that are generated from antibody gene sequences allow a functional analysis of the repertoire. This review discusses what has been learned thus far from analysis of diverse pathogen-specific memory B cell compartments and describes major differences in their repertoires. Such information may illuminate ways to advance the goal of improving vaccine and therapeutic antibody design.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Memoria Inmunológica , Células Plasmáticas/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/metabolismo , Antígenos/inmunología , Subgrupos de Linfocitos B/metabolismo , Linfocitos B/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Humoral/inmunología , Células Plasmáticas/metabolismo
19.
Data Brief ; 9: 213-9, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27656675

RESUMEN

Previously published studies revealed that variation in expression of the DNA-binding protein ARID3a in B lymphocytes from patients with systemic lupus erythematosus (SLE) correlated with levels of disease activity ("Disease activity in systemic lupus erythematosus correlates with expression of the transcription factor AT-rich-interactive domain 3A" (J.M. Ward, K. Rose, C. Montgomery, I. Adrianto, J.A. James, J.T. Merrill et al., 2014) [1]). The data presented here compare DNA methylation patterns from SLE peripheral blood mononuclear cells obtained from samples with high numbers of ARID3a expressing B cells (ARID3a(H)) versus SLE samples with normal numbers of ARID3a(+) B cells (ARID3a(N)). The methylation data is available at the gene expression omnibus (GEO) repository, "Gene Expression Omnibus: NCBI gene expression and hybridization array data repository" (R. Edgar, M. Domrachev, A.E. Lash, 2002) [2]. Isolated B cells from SLE ARID3a(H) and ARID3a(N) B samples were also evaluated via qRT-PCR for Type I interferon (IFN) signature and pathway gene expression levels by qRT-PCR. Similarly, healthy control B cells and B cells stimulated to express ARID3a with the TLR agonist, CpG, were also compared via qRT-PCR. Primers designed to detect 6 IFNa subtype mRNAs were tested in 4 IFNa, Epstein-Barr Virus-transformed B cell lines ("Reduced interferon-alpha production by Epstein-Barr virus transformed B-lymphoblastoid cell lines and lectin-stimulated lymphocytes in congenital dyserythropoietic anemia type I" (S.H. Wickramasinghe, R. Hasan, J. Smythe, 1997) [3]). The data in this article support the publication, "Human effector B lymphocytes express ARID3a and secrete interferon alpha" (J.M. Ward, M.L. Ratliff, M.G. Dozmorov, G. Wiley, J.M. Guthridge, P.M. Gaffney, J.A. James, C.F. Webb, 2016) [4].

20.
Antibodies (Basel) ; 4: 354-368, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28580178

RESUMEN

Earlier studies showed that the DNA-binding protein, Bright/ARID3a bound to a subset of human and mouse immunoglobulin heavy chain promoters where it enhanced expression. Indeed, mice with transgenic expression of ARID3a in all B lymphocytes have expanded MZ B cells and produce anti-nuclear antibodies (ANAs). Consistent with our findings in mice, we observed that human systemic lupus erythematosus (SLE) patients had expanded numbers of peripheral blood ARID3a+ B cells that were associated with increased disease activity (p = 0.0038). We hypothesized that ARID3a+ naïve B cells would eventually produce autoantibodies, explaining associations between ARID3a expression and disease activity in lupus. Unlike healthy controls, ARID3a was expressed in the naïve B cell population in SLE patients, and we hypothesized that these might represent expansions of autoreactive cells. Therefore, monoclonal antibodies were generated from single-sorted naïve B cells derived from patients with normal (ARID3aN) and high (ARID3aH) numbers of ARID3a+ B cells. We found that ARID3a expression did not correlate with autoantibody expression. Furthermore, measures of antigen specificities of autoreactive antibodies did not reveal skewing toward particular proteins. These data suggest that the association of increased disease activity in SLE with numbers of ARID3a+ B lymphocytes may be mediated by an antibody-independent mechanism.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA