Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Clin Immunol ; 226: 108716, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33774179

RESUMEN

Lung inflammation and damage is prominent in people infected with SARS-Cov-2 and a major determinant of morbidity and mortality. We report the deposition of complement components in the lungs of people who succumbed to COVID-19 consistent with the activation of the classical and the alternative pathways. Our study provides strong rationale for the expansion of trials involving the use of complement inhibitors to treat patients with COVID-19.


Asunto(s)
COVID-19/inmunología , Activación de Complemento/inmunología , Vía Alternativa del Complemento/inmunología , Lesión Pulmonar/inmunología , Anciano , Anciano de 80 o más Años , COVID-19/complicaciones , Inactivadores del Complemento/farmacología , Inactivadores del Complemento/uso terapéutico , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Humanos , Inmunohistoquímica , Pulmón/diagnóstico por imagen , Pulmón/inmunología , Pulmón/patología , Lesión Pulmonar/complicaciones , Lesión Pulmonar/patología , Lesión Pulmonar/virología , Masculino , Persona de Mediana Edad
2.
Clin Immunol ; 229: 108795, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34252574

RESUMEN

Acute and chronic kidney failure is common in hospitalized patients with COVID-19, yet the mechanism of injury and predisposing factors remain poorly understood. We investigated the role of complement activation by determining the levels of deposited complement components (C1q, C3, FH, C5b-9) and immunoglobulin along with the expression levels of the injury-associated molecules spleen tyrosine kinase (Syk), mucin-1 (MUC1) and calcium/calmodulin-dependent protein kinase IV (CaMK4) in the kidney tissues of people who succumbed to COVID-19. We report increased deposition of C1q, C3, C5b-9, total immunoglobulin, and high expression levels of Syk, MUC1 and CaMK4 in the kidneys of COVID-19 patients. Our study provides strong rationale for the expansion of trials involving the use of inhibitors of these molecules, in particular C1q, C3, Syk, MUC1 and CaMK4 to treat patients with COVID-19.


Asunto(s)
COVID-19/metabolismo , Proteínas del Sistema Complemento/metabolismo , Riñón/metabolismo , Mucina-1/metabolismo , SARS-CoV-2 , Quinasa Syk/metabolismo , Anciano , Anciano de 80 o más Años , COVID-19/patología , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/metabolismo , Proteínas del Sistema Complemento/genética , Resultado Fatal , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Mucina-1/genética , Quinasa Syk/genética
3.
Rheumatology (Oxford) ; 60(1): 420-429, 2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-32810232

RESUMEN

OBJECTIVE: CD4 T helper 1 (Th1) cells producing IFN-γ contribute to inflammatory responses in the pathogenesis of SLE and lupus nephritis. Moreover, elevated serum type II IFN levels precede the appearance of type I IFNs and autoantibodies in patient years before clinical diagnosis. However, the molecules and mechanisms that control this inflammatory response in SLE remain unclear. Serine/arginine-rich splicing factor 1 (SRSF1) is decreased in T cells from SLE patients, and restrains T cell hyperactivity and systemic autoimmunity. Our objective here was to evaluate the role of SRSF1 in IFN-γ production, Th1 differentiation and experimental nephritis. METHODS: T cell-conditional Srsf1-knockout mice were used to study nephrotoxic serum-induced nephritis and evaluate IFN-γ production and Th1 differentiation by flow cytometry. RNA sequencing was used to assess transcriptomics profiles. RhoH was silenced by siRNA transfections in human T cells by electroporation. RhoH and SRSF1 protein levels were assessed by immunoblots. RESULTS: Deletion of Srsf1 in T cells led to increased Th1 differentiation and exacerbated nephrotoxic serum nephritis. The expression levels of RhoH are decreased in Srsf1-deficient T cells, and silencing RhoH in human T cells leads to increased production of IFN-γ. Furthermore, RhoH expression was decreased and directly correlated with SRSF1 in T cells from SLE patients. CONCLUSION: Our study uncovers a previously unrecognized role of SRSF1 in restraining IFN-γ production and Th1 differentiation through the control of RhoH. Reduced expression of SRSF1 may contribute to pathogenesis of autoimmune-related nephritis through these molecular mechanisms.


Asunto(s)
Diferenciación Celular/fisiología , Interferón gamma/metabolismo , Nefritis Lúpica/genética , Factores de Empalme Serina-Arginina/genética , Células TH1/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Citometría de Flujo , Humanos , Nefritis Lúpica/inmunología , Nefritis Lúpica/metabolismo , Ratones , Ratones Noqueados , Factores de Empalme Serina-Arginina/metabolismo , Células TH1/inmunología
4.
Rheumatology (Oxford) ; 59(8): 2146-2155, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32206811

RESUMEN

OBJECTIVE: Lymphopenia is a frequent clinical manifestation and risk factor for infections in SLE, but the underlying mechanisms are not fully understood. We previously identified novel roles for the RNA-binding protein serine arginine-rich splicing factor 1 (SRSF1) in the control of genes involved in signalling and cytokine production in human T cells. SRSF1 is decreased in T cells from patients with SLE and associates with severe disease. Because SRSF1 controls the expression of apoptosis-related genes, we hypothesized that SRSF1 controls T cell homeostasis and, when reduced, leads to lymphopenia. METHODS: We evaluated SRSF1 expression in T cells from SLE patients by immunoblots and analysed its correlation with clinical parameters. T cell conditional Srsf1 knockout mice were used to evaluate lymphoid cells and apoptosis by flow cytometry. Quantitative PCR and immunoblots were used to assess Bcl-xL mRNA and protein expression. SRSF1 overexpression was performed by transient transfections by electroporation. RESULTS: We found that low SRSF1 levels correlated with lymphopenia in SLE patients. Selective deletion of Srsf1 in T cells in mice led to T cell lymphopenia, with increased apoptosis and decreased expression of the anti-apoptotic Bcl-xL. Lower SRSF1 expression correlated with low Bcl-xL levels in T cells and lower Bcl-xL levels associated with lymphopenia in SLE patients. Importantly, overexpression of SRSF1 rescued survival of T cells from patients with SLE. CONCLUSION: Our studies uncovered a previously unrecognized role for SRSF1 in the control of T cell homeostasis and its reduced expression as a molecular defect that contributes to lymphopenia in systemic autoimmunity.


Asunto(s)
Homeostasis/fisiología , Lupus Eritematoso Sistémico/metabolismo , Linfopenia/metabolismo , Factores de Empalme Serina-Arginina/metabolismo , Linfocitos T/metabolismo , Adulto , Animales , Femenino , Humanos , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/genética , Activación de Linfocitos/fisiología , Linfopenia/genética , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Factores de Empalme Serina-Arginina/sangre , Factores de Empalme Serina-Arginina/genética , Adulto Joven , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
5.
J Immunol ; 200(9): 3077-3086, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29602774

RESUMEN

Cytotoxic function and cytokine profile of NK cells are compromised in patients with systemic lupus erythematosus (SLE). CD3ζ, an important molecule for NK cell activation, is downregulated in SLE T cells and contributes to their altered function. However, little is known about the role of CD3ζ in SLE NK cells. We studied CD3ζ levels and its contribution to cytotoxic, degranulation, and cytokine production capacity of NK cells from patients with SLE. Furthermore, we studied the human NK cell line, NKL, in which manipulation of CD3ζ levels was achieved using small interfering RNA and NK cells from Rag2 mice deficient in CD3ζ. We found reduced CD3ζ expression in NK cells from SLE patients independent of disease activity. Downregulation of CD3ζ expression in NK cells is mediated, at least in part, by Caspase 3, the activity of which is higher in NK cells from patients with SLE compared with NK cells from healthy donors. CD3ζ levels correlated inversely with natural cytotoxicity and the percentage of cells capable of producing the proinflammatory cytokines IFN-γ and TNF. In contrast, CD3ζ levels showed a direct correlation with levels of Ab-dependent cellular cytotoxicity. Experiments performed in CD3ζ-silenced NKL and CD3ζ-deficient NK cells from Rag2 mice confirmed the dependence of NK cell function on CD3ζ levels. Our results demonstrate a differential role for CD3ζ in natural cytotoxicity and Ab-dependent cellular cytotoxicity. We conclude that downregulated CD3ζ confers a proinflammatory phenotype to SLE NK cells and contributes to their altered function in patients with SLE.


Asunto(s)
Complejo CD3/inmunología , Células Asesinas Naturales/inmunología , Lupus Eritematoso Sistémico/inmunología , Animales , Complejo CD3/metabolismo , Regulación hacia Abajo , Femenino , Humanos , Activación de Linfocitos/inmunología , Ratones , Fenotipo
6.
J Immunol ; 198(11): 4268-4276, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28438897

RESUMEN

Systemic lupus erythematosus (SLE) damages multiple organs by producing various autoantibodies. In this study, we report that decreased microRNA (miR)-200a-3p causes IL-2 hypoproduction through zinc finger E-box binding homeobox (ZEB)1 and C-terminal binding protein 2 (CtBP2) in a lupus-prone mouse. First, we performed RNA sequencing to identify candidate microRNAs and mRNAs involved in the pathogenesis of SLE. We found that miR-200a-3p was significantly downregulated, whereas its putative targets, ZEB2 and CtBP2, were upregulated in CD4+ T cells from MRL/lpr-Tnfrsf6lpr mice compared with C57BL/6J mice. ZEB1 and ZEB2 comprise the ZEB family and suppress various genes, including IL-2 by recruiting CtBP2. IL-2 plays a critical role in immune tolerance, and insufficient IL-2 production upon stimulation has been recognized in SLE pathogenesis. Therefore, we hypothesized that decreased miR-200a-3p causes IL-2 deficit through the ZEB1-CtBP2 and/or ZEB2-CtBP2 complex in SLE CD4+ T cells. Overexpression of miR-200a-3p induced IL-2 production by downregulating ZEB1, ZEB2, and CtBP2 in EL4 cell lines. We further revealed that miR-200a-3p promotes IL-2 expression by reducing the binding of suppressive ZEB1-CtBP2 and ZEB2-CtBP2 complexes on negative regulatory element A in the IL-2 promoter in EL4 cells. Interestingly, the ZEB1-CtBP2 complex on negative regulatory element A was significantly upregulated after PMA/ionomycin stimulation in lupus CD4+ T cells. Our studies have revealed a new epigenetic pathway in the control of IL-2 production in SLE whereby low levels of miR-200a-3p accumulate the binding of the ZEB1-CtBP2 complex to the IL-2 promoter and suppress IL-2 production.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Interleucina-2/biosíntesis , Interleucina-2/genética , Lupus Eritematoso Sistémico/inmunología , MicroARNs/genética , Fosfoproteínas/genética , Linfocitos T/inmunología , Oxidorreductasas de Alcohol , Animales , Línea Celular , Proteínas Co-Represoras , Proteínas de Unión al ADN/metabolismo , Interleucina-2/inmunología , Lupus Eritematoso Sistémico/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Fosfoproteínas/metabolismo , Linfocitos T/patología , Activación Transcripcional , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
7.
Proc Natl Acad Sci U S A ; 110(5): 1845-50, 2013 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-23319613

RESUMEN

T cells from patients with systemic lupus erythematosus (SLE) produce insufficient amounts of the vital cytokine IL-2. We previously showed that SLE T cells express decreased levels of the T-cell receptor-CD3ζ chain and forced expression of CD3ζ into SLE T cells restores IL-2 production. We recently showed that the serine arginine protein splicing factor 2/alternative splicing factor (SF2/ASF) enhances the expression of CD3ζ chain by limiting the production of an unstable splice variant. Here we demonstrate that SF2/ASF levels are decreased in patients with SLE and more so in those with active disease. More importantly, we reveal a function of SF2/ASF, independent of T-cell receptor/CD3 signaling, whereby it is recruited to the IL-2 promoter, increases transcriptional activity, and enhances IL-2 production in SLE T cells. Our results demonstrate that SF2/ASF regulates IL-2 production and that decreased SF2/ASF expression in SLE T cells contributes to deficient IL-2 production.


Asunto(s)
Interleucina-2/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/metabolismo , Linfocitos T/metabolismo , Adulto , Femenino , Humanos , Immunoblotting , Interleucina-2/genética , Lupus Eritematoso Sistémico/genética , Masculino , Persona de Mediana Edad , Proteínas Nucleares/genética , Proteínas de Unión al ARN/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Empalme Serina-Arginina , Transducción de Señal , Activación Transcripcional , Adulto Joven
8.
J Biol Chem ; 289(7): 4126-34, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24368769

RESUMEN

T cells from patients with systemic lupus erythematosus (SLE) exhibit reduced expression of the critical T cell receptor (TCR)-associated CD3ζ signaling chain and are poor producers of the vital cytokine IL-2. By oligonucleotide pulldown and mass spectrometry discovery approaches, we identified the splicing regulator serine/arginine-rich splicing factor (SRSF) 1 or splicing factor 2/alternative splicing factor (SF2/ASF) to be important in the expression of CD3ζ chain. Importantly, increases in the expression of SRSF1 rescued IL-2 production in T cells from patients with SLE. In this study, we investigated the regulation of SRSF1 expression in resting and activated human T cells. We found that T cell stimulation induced a rapid and significant increase in mRNA expression of SRSF1; however, protein expression levels did not correlate with this increase. Co-engagement of CD28 induced a similar mRNA induction and reduction in protein levels. Proteasomal but not lysosomal degradation was involved in this down-regulation as evidenced by blocking with specific inhibitors MG132 and bafilomycin, respectively. Immunoprecipitation studies showed increased ubiquitination of SRSF1 in activated T cells. Interestingly, T cells from patients with SLE showed increased ubiquitination of SRSF1 when compared with those from healthy individuals. Our results demonstrate a novel mechanism of regulation of the splicing factor SRSF1 in human T cells and a potential molecular mechanism that controls its expression in SLE.


Asunto(s)
Regulación de la Expresión Génica , Lupus Eritematoso Sistémico/metabolismo , Activación de Linfocitos , Proteínas Nucleares/biosíntesis , Proteolisis , Proteínas de Unión al ARN/biosíntesis , Linfocitos T/metabolismo , Antígenos CD28/biosíntesis , Antígenos CD28/genética , Antígenos CD28/inmunología , Complejo CD3/biosíntesis , Complejo CD3/genética , Complejo CD3/inmunología , Femenino , Humanos , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Masculino , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/inmunología , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Mensajero/inmunología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Factores de Empalme Serina-Arginina , Linfocitos T/inmunología , Linfocitos T/patología , Ubiquitinación
10.
Mol Med ; 18: 370-8, 2012 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-22281835

RESUMEN

Systemic lupus erythematosus (SLE) is an autoimmune disease with a complex multifactorial pathogenesis. T lymphocytes play a critical role in disease pathogenesis and display abnormal gene expression and poor interleukin (IL)-2 production. We previously showed that the expression of the transcriptional repressor cyclic AMP response element modulator α (CREMα) is increased in SLE T cells and contributes to reduced IL-2 production. Although estrogen is implicated in the onset and exacerbation of SLE, the precise nature of molecular events regulated by estrogen in immune cell function is not well understood. Here, we asked whether estrogen regulates the expression of CREMα in human T lymphocytes. We show that exposure of human T cells to 17-ß-estradiol leads to a dose-dependent increase in CREMα mRNA expression, and this increase appears to be mediated through the estrogen receptors α and ß. We show that the increased expression of CREMα is due to increased transcriptional activity of the CREM promoter and is mediated by increased expression and binding of the Sp1 transcriptional activator. We further show that estrogen treatment leads to a dose-dependent decrease in IL-2 mRNA and cytokine production by T cells. Finally, the effect of ß-estradiol on CREMα is observed more frequently in T cells from women than from men. We conclude that estrogen can modulate the expression of CREMα and lead to IL-2 suppression in human T lymphocytes, thus revealing a molecular link between hormones and the immune system in SLE.


Asunto(s)
Modulador del Elemento de Respuesta al AMP Cíclico/biosíntesis , Estrógenos/metabolismo , Interleucina-2/metabolismo , Linfocitos T/metabolismo , Adulto , Modulador del Elemento de Respuesta al AMP Cíclico/genética , Regulación hacia Abajo , Femenino , Humanos , Lupus Eritematoso Sistémico/metabolismo , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas , Factor de Transcripción Sp1/metabolismo , Regulación hacia Arriba/efectos de los fármacos
11.
Mol Immunol ; 152: 140-152, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36368121

RESUMEN

Regulatory T cells (Tregs) are vital for maintaining immune self-tolerance, and their impaired function leads to autoimmune disease. Mutations in FoxP3, the master transcriptional regulator of Tregs, leads to immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome in humans and the early lethal "scurfy" phenotype with multi-organ autoimmune disease in mice. We recently identified serine/arginine-rich splicing factor 1 (SRSF1) as an indispensable regulator of Treg homeostasis and function. Intriguingly, Treg-conditional SRSF1-deficient mice exhibit early lethal systemic autoimmunity with multi-organ inflammation reminiscent of the scurfy mice. Importantly, SRSF1 is decreased in T cells from patients with the autoimmune disease systemic lupus erythematosus (SLE), and low SRSF1 levels inversely correlate with disease severity. Given that the Treg-specific deficiency of SRSF1 causes similarly profound autoimmune disease outcomes in mice as the deficiency/mutation in FoxP3, we aimed to evaluate the genes and molecular pathways controlled by these two indispensable regulatory proteins. We performed comparative bioinformatic analyses of transcriptomic profiles of Tregs from Srsf1-knockout mice and two Foxp3 mutant mice--the FoxP3-deficient ΔFoxp3 and the Foxp3 M370I mutant mice. We identified 132 differentially expressed genes (DEGs) unique to Srsf1-ko Tregs, 503 DEGs unique to Foxp3 M370I Tregs, and 1367 DEGs unique to ΔFoxp3 Tregs. Gene set enrichment and pathway analysis of DEGs unique to Srsf1-ko Tregs indicate that SRSF1 controls cytokine and immune response pathways. Conversely, FoxP3 controls pathways involved in DNA replication and cell cycle. Besides the distinct gene signatures, we identified only 30 shared genes between all three Treg mutants, mostly contributing to cytokine and immune defense pathways. Prominent genes included the chemokines CXCR6 and CCL1 and the checkpoint inhibitors FASLG and PDCD1. Thus, we demonstrate that SRSF1 and FoxP3 control common and distinct molecular pathways implicated in autoimmunity. Our analyses suggest that SRSF1 controls crucial immune functions in Tregs contributing to immune tolerance, and perturbations in its levels lead to systemic autoimmunity via mechanisms that are largely distinct from FoxP3.


Asunto(s)
Diabetes Mellitus Tipo 1 , Lupus Eritematoso Sistémico , Factores de Empalme Serina-Arginina , Animales , Humanos , Ratones , Citocinas/metabolismo , Factores de Transcripción Forkhead , Lupus Eritematoso Sistémico/metabolismo , Ratones Noqueados , Factores de Empalme de ARN/metabolismo , Factores de Empalme Serina-Arginina/genética , Linfocitos T Reguladores
12.
Mol Immunol ; 141: 94-103, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34839165

RESUMEN

Systemic autoimmune diseases are characterized by hyperactive effector T cells (Teffs), aberrant cytokines and chemokines, and dysfunctional regulatory T cells (Tregs). We previously uncovered new roles for serine/arginine-rich splicing factor 1 (SRSF1) in the control of genes involved in T cell signaling and cytokine production in human T cells. SRSF1 levels are decreased in T cells from patients with systemic lupus erythematosus (SLE), and low levels correlate with severe disease. Moreover, T cell-conditional Srsf1-deficient mice recapitulate the autoimmune phenotype, exhibiting CD4 T cell hyperactivity, dysfunctional Tregs, systemic autoimmunity, and tissue inflammation. However, the role of SRSF1 in controlling molecular programs in Teffs and Tregs and how these pathways are implicated in autoimmunity is not known. Here, by comparative bioinformatics analysis, we demonstrate that SRSF1 controls largely distinct gene programs in Tregs and Teffs in vivo. SRSF1 regulates 189 differentially expressed genes (DEGs) unique to Tregs, 582 DEGs unique to Teffs, and 29 DEGs shared between both. Shared genes included IL-17A, IL-17F, CSF1, CXCL10, and CXCR4, and were highly enriched for inflammatory response and cytokine-cytokine receptor interaction pathways. SRSF1 controls distinct pathways in Tregs, which include chemokine signaling and immune cell differentiation, compared with pathways in Teffs, which include cytokine production, T cell homeostasis, and activation. We identified putative mRNA binding targets of SRSF1 which include CSF1, CXCL10, and IL-17F. Finally, comparisons with transcriptomics profiles from lupus-prone MRL/lpr mice reveal that SRSF1 controls genes and pathways implicated in autoimmune disease. The target genes of SRSF1 and putative binding targets we discovered, have known roles in systemic autoimmunity. Our findings suggest that SRSF1 controls distinct molecular pathways in Tregs and Teffs and aberrant SRSF1 levels may contribute to their dysfunction and immunopathogenesis of systemic autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Factores de Empalme de ARN/inmunología , Factores de Empalme Serina-Arginina/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Citocinas/inmunología , Inflamación/inmunología , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Noqueados , ARN Mensajero/inmunología , Transducción de Señal/inmunología , Transcriptoma/inmunología
13.
Front Immunol ; 13: 906355, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36189299

RESUMEN

Cytotoxic CD8 T cells are crucial for the host antigen-specific immune response to viral pathogens. Here we report the identification of an essential role for the serine/arginine-rich splicing factor (SRSF) 1 in CD8 T cell homeostasis and function. Specifically, SRSF1 is necessary for the maintenance of normal CD8 T lymphocyte numbers in the lymphoid compartment, and for the proliferative capacity and cytotoxic function of CD8 T cells. Furthermore, SRSF1 is required for antigen-specific IFN-γ cytokine responses in a viral infection challenge in mice. Transcriptomics analyses of Srsf1-deficient T cells reveal that SRSF1 controls proliferation, MAP kinase signaling and IFN signaling pathways. Mechanistically, SRSF1 controls the expression and activity of the Mnk2/p38-MAPK axis at the molecular level. Our findings reveal previously unrecognized roles for SRSF1 in the physiology and function of cytotoxic CD8 T lymphocytes and a potential molecular mechanism in viral immunopathogenesis.


Asunto(s)
Linfocitos T CD8-positivos , Citocinas , Factores de Empalme Serina-Arginina/inmunología , Animales , Arginina , Linfocitos T CD8-positivos/metabolismo , Citocinas/metabolismo , Inmunidad , Ratones , Factores de Empalme de ARN , Serina , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo
14.
J Biol Chem ; 285(17): 12490-6, 2010 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-20118245

RESUMEN

T cells from patients with systemic lupus erythematosus express decreased levels of the T cell receptor-associated CD3 zeta chain, a feature directly linked to their aberrant function. The decrease in CD3zeta protein expression is in part due to decreased levels of functional wild type isoform of the 3'-untranslated region (UTR) of CD3zeta mRNA with concomitant increased levels of an unstable alternatively spliced isoform. In order to identify factors involved in the post-transcriptional regulation of CD3zeta, we performed mass spectrometric analysis of Jurkat T cell nuclear proteins "pulled down" by a CD3zeta 3'-UTR oligonucleotide, which identified the splicing protein alternative splicing factor/splicing factor 2 (ASF/SF2). We show for the first time that ASF/SF2 binds specifically to the 3'-UTR of CD3zeta and regulates expression of CD3zeta protein by limiting the production of the alternatively spliced isoform. During activation of human T cells, an increase in the wild type CD3zeta mRNA is associated with increased expression of ASF/SF2. Finally, we show a significant correlation between ASF/SF2 and CD3zeta protein levels in T cells from systemic lupus erythematosus patients. Thus, our results identify ASF/SF2 as a novel factor in the regulation of alternative splicing of the 3'-UTR of CD3zeta and protein expression in human T cells.


Asunto(s)
Empalme Alternativo , Complejo CD3/biosíntesis , Lupus Eritematoso Sistémico/metabolismo , Proteínas Nucleares/metabolismo , Linfocitos T/metabolismo , Regiones no Traducidas 3' , Femenino , Humanos , Células Jurkat , Masculino , Isoformas de Proteínas/biosíntesis , Proteínas de Unión al ARN , Factores de Empalme Serina-Arginina
15.
Cell Rep ; 36(1): 109339, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34233194

RESUMEN

The ability of regulatory T (Treg) cells to control the immune response and limit the development of autoimmune diseases is determined by distinct molecular processes, which are not fully understood. We show here that serine/arginine-rich splicing factor 1 (SRSF1), which is decreased in T cells from patients with systemic lupus erythematosus, is necessary for the homeostasis and proper function of Treg cells, because its conditional absence in these cells leads to profound autoimmunity and organ inflammation by elevating the glycolytic metabolism and mTORC1 activity and the production of proinflammatory cytokines. Our data reveal a molecular mechanism that controls Treg cell plasticity and offer insights into the pathogenesis of autoimmune disease.


Asunto(s)
Homeostasis , Factores de Empalme Serina-Arginina/metabolismo , Linfocitos T/metabolismo , Animales , Plasticidad de la Célula , Supervivencia Celular , Eliminación de Gen , Glucólisis , Heterocigoto , Inflamación/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Factores de Empalme Serina-Arginina/deficiencia , Sirolimus/farmacología , Linfocitos T Reguladores/inmunología , Transcriptoma/genética
16.
Arthritis Rheumatol ; 73(11): 2052-2058, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33982889

RESUMEN

OBJECTIVE: Autoimmune diseases affect women disproportionately more than men. Estrogen is implicated in immune cell dysfunction, yet its precise molecular roles are not fully known. We recently identified new roles for serine/arginine-rich splicing factor 1 (SRSF1) in T cell function and autoimmunity. SRSF1 levels are decreased in T cells from patients with systemic lupus erythematosus (SLE) and are associated with active disease and comorbidity. However, the molecular mechanisms that control SRSF1 expression are unknown. Srsf1 messenger RNA (mRNA) has a long 3'-untranslated region (3'-UTR), suggesting posttranscriptional control. This study was undertaken to investigate the role of estrogen and posttranscriptional mechanisms of SRSF1 regulation in T cells and SLE. METHODS: In silico bioinformatics analysis of Srsf1-3'-UTR revealed multiple microRNA (miRNA; miR)-binding sites. Additional screening and literature searches narrowed down hsa-miR-10b-5p for further study. Peripheral blood T cells from healthy individuals and SLE patients were evaluated for mRNA and miRNA expression by quantitative reverse transcription-polymerase chain reaction, and SRSF1 protein levels were assessed by immunoblotting. T cells were cultured with ß-estradiol, and transient transfections were used to overexpress miRNAs. Luciferase assays were used to measure 3'-UTR activity. RESULTS: We demonstrated that estrogen increased hsa-miR-10b-5p expression in human T cells, and hsa-miR-10b-5p down-regulated SRSF1 protein expression. Mechanistically, hsa-mir-10b-5p regulated SRSF1 posttranscriptionally via control of its 3'-UTR activity. Importantly, hsa-miR-10b-5p expression levels were elevated in T cells from healthy women compared to healthy men and also elevated in T cells from SLE patients. CONCLUSION: We identified a previously unrecognized molecular link between estrogen and gene regulation in immune cells, with potential relevance to systemic autoimmune disease.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Estradiol/farmacología , Lupus Eritematoso Sistémico/metabolismo , MicroARNs/metabolismo , Factores de Empalme Serina-Arginina/metabolismo , Linfocitos T/efectos de los fármacos , Adulto , Anciano , Simulación por Computador , Femenino , Células HEK293 , Humanos , Lupus Eritematoso Sistémico/genética , Activación de Linfocitos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Factores de Empalme Serina-Arginina/genética , Linfocitos T/metabolismo , Adulto Joven
17.
Front Immunol ; 11: 589474, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193418

RESUMEN

The SARS-CoV-2 novel coronavirus has caused the COVID-19 pandemic with over 35 million cases and over a million deaths worldwide as of early October 2020. The populations most affected are the elderly and especially those with underlying comorbidities. In terms of race and ethnicity, black and hispanic populations are affected at disproportionately higher rates. Individuals with underlying conditions that cause an immune-compromised state are considered vulnerable to this infection. The immune response is an important determinant in viral infections including coronaviruses, not only in the antiviral defense but also in the disease progression, severity, and clinical outcomes of COVID-19. Systemic lupus erythematosus is a chronic autoimmune disease which also disproportionately afflicts black and hispanic populations. In lupus patients, an aberrant immune response is characterized by the presence of circulating autoantibodies, lymphopenia, aberrant T cells, and proinflammatory cytokines along with defective regulatory mechanisms, leading to immune-mediated damage to tissues. Lupus patients are often treated with immune-suppressants and therefore are immune-compromised and more susceptible to infections and may be vulnerable to coronavirus infection. While the anti-viral immune response is important to protect from coronavirus infection, an uncontrolled proinflammatory cytokine response can lead to cytokine storm which causes damage to the lungs and other organs, causing significant morbidity and mortality. Better understanding of the underlying immune response and therapeutic strategies in lupus and COVID-19 is important to guide management of this deadly infectious disease in the context of lupus and vice-versa.


Asunto(s)
COVID-19/inmunología , Lupus Eritematoso Sistémico/inmunología , SARS-CoV-2/fisiología , Animales , COVID-19/genética , COVID-19/virología , Humanos , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/genética , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Tratamiento Farmacológico de COVID-19
18.
Front Immunol ; 11: 2147, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32983176

RESUMEN

The novel severe acute respiratory syndrome coronavirus 2, the cause of the coronavirus disease 2019 (COVID-19) pandemic, has ravaged the world, with over 22 million total cases and over 770,000 deaths worldwide as of August 18, 2020. While the elderly are most severely affected, implicating an age bias, a striking factor in the demographics of this deadly disease is the gender bias, with higher numbers of cases, greater disease severity, and higher death rates among men than women across the lifespan. While pre-existing comorbidities and social, behavioral, and lifestyle factors contribute to this bias, biological factors underlying the host immune response may be crucial contributors. Women mount stronger immune responses to infections and vaccinations and outlive men. Sex-based biological factors underlying the immune response are therefore important determinants of susceptibility to infections, disease outcomes, and mortality. Despite this, gender is a profoundly understudied and often overlooked variable in research related to the immune response and infectious diseases, and it is largely ignored in drug and vaccine clinical trials. Understanding these factors will not only help better understand the pathogenesis of COVID-19, but it will also guide the design of effective therapies and vaccine strategies for gender-based personalized medicine. This review focuses on sex-based differences in genes, sex hormones, and the microbiome underlying the host immune response and their relevance to infections with a focus on coronaviruses.


Asunto(s)
Betacoronavirus/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/mortalidad , Interacciones Huésped-Patógeno/inmunología , Neumonía Viral/inmunología , Neumonía Viral/mortalidad , Inmunidad Adaptativa/genética , COVID-19 , Infecciones por Coronavirus/virología , Femenino , Predisposición Genética a la Enfermedad , Hormonas Esteroides Gonadales/inmunología , Interacciones Huésped-Patógeno/genética , Humanos , Inmunidad Innata/genética , Masculino , Microbiota/inmunología , Pandemias , Neumonía Viral/virología , SARS-CoV-2 , Factores Sexuales
19.
Front Physiol ; 11: 571416, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33510644

RESUMEN

The novel coronavirus severe acute respiratory syndrome coronavirus 2 causing the Coronavirus disease (COVID-19) pandemic has ravaged the world with over 72 million total cases and over 1.6 million deaths worldwide as of early December 2020. An overwhelming preponderance of cases and deaths is observed within the elderly population, and especially in those with pre-existing conditions and comorbidities. Aging causes numerous biological changes in the immune system, which are linked to age-related illnesses and susceptibility to infectious diseases. Age-related changes influence the host immune response and therefore not only weaken the ability to fight respiratory infections but also to mount effective responses to vaccines. Immunosenescence and inflamm-aging are considered key features of the aging immune system wherein accumulation of senescent immune cells contribute to its decline and simultaneously increased inflammatory phenotypes cause immune dysfunction. Age-related quantitative and qualitative changes in the immune system affect cells and soluble mediators of both the innate and adaptive immune responses within lymphoid and non-lymphoid peripheral tissues. These changes determine not only the susceptibility to infections, but also disease progression and clinical outcomes thereafter. Furthermore, the response to therapeutics and the immune response to vaccines are influenced by age-related changes within the immune system. Therefore, better understanding of the pathophysiology of aging and the immune response will not only help understand age-related diseases but also guide targeted management strategies for deadly infectious diseases like COVID-19.

20.
Nat Commun ; 11(1): 2859, 2020 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-32503973

RESUMEN

Mature double negative (DN) T cells are a population of αß T cells that lack CD4 and CD8 coreceptors and contribute to systemic lupus erythematosus (SLE). The splenic marginal zone macrophages (MZMs) are important for establishing immune tolerance, and loss of their number or function contributes to the progression of SLE. Here we show that loss of MZMs impairs the tolerogenic clearance of apoptotic cells and alters the serum cytokine profile, which in turn provokes the generation of DN T cells from self-reactive CD8+ T cells. Increased Ki67 expression, narrowed TCR V-beta repertoire usage and diluted T-cell receptor excision circles confirm that DN T cells from lupus-prone mice and patients with SLE undergo clonal proliferation and expansion in a self-antigen dependent manner, which supports the shared mechanisms for their generation. Collectively, our results provide a link between the loss of MZMs and the expansion of DN T cells, and indicate possible strategies to prevent the development of SLE.


Asunto(s)
Autoantígenos/inmunología , Interleucina-17/metabolismo , Lupus Eritematoso Sistémico/inmunología , Subgrupos de Linfocitos T/inmunología , Traslado Adoptivo , Animales , Autoantígenos/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Tolerancia Inmunológica , Antígeno Ki-67/inmunología , Antígeno Ki-67/metabolismo , Lupus Eritematoso Sistémico/sangre , Macrófagos/inmunología , Ratones , Ratones Noqueados , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Subgrupos de Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA