Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Circulation ; 138(22): 2513-2526, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-29997115

RESUMEN

BACKGROUND: Atherosclerotic cardiovascular disease is a chronic inflammatory process initiated when cholesterol-carrying low-density lipoprotein (LDL) is retained in the arterial wall. CD4+ T cells, some of which recognize peptide components of LDL as antigen, are recruited to the forming lesion, resulting in T-cell activation. Although these T cells are thought to be proatherogenic, LDL immunization reduces disease in experimental animals. These seemingly contradictory findings have hampered the development of immune-based cardiovascular therapy. The present study was designed to clarify how activation of LDL-reactive T cells impacts on metabolism and vascular pathobiology. METHODS: We have developed a T-cell receptor-transgenic mouse model to characterize the effects of immune reactions against LDL. Through adoptive cell transfers and cross-breeding to hypercholesterolemic mice expressing the antigenic human LDL protein apolipoprotein B-100, we evaluate the effects on atherosclerosis. RESULTS: A subpopulation of LDL-reactive T cells survived clonal selection in the thymus, developed into T follicular helper cells in lymphoid tissues on antigen recognition, and promoted B-cell activation. This led to production of anti-LDL immunoglobulin G antibodies that enhanced LDL clearance through immune complex formation. Furthermore, the cellular immune response to LDL was associated with increased cholesterol excretion in feces and with reduced vascular inflammation. CONCLUSIONS: These data show that anti-LDL immunoreactivity evokes 3 atheroprotective mechanisms: antibody-dependent LDL clearance, increased cholesterol excretion, and reduced vascular inflammation.


Asunto(s)
Aterosclerosis/prevención & control , Linfocitos T CD4-Positivos/inmunología , Colesterol/sangre , Lipoproteínas LDL/inmunología , Animales , Anticuerpos/inmunología , Apolipoproteína B-100/sangre , Apolipoproteínas E , Aterosclerosis/patología , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Modelos Animales de Enfermedad , Lipoproteínas LDL/administración & dosificación , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo
2.
Circ Res ; 120(11): 1740-1753, 2017 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-28420668

RESUMEN

RATIONALE: The liver is the central organ that responds to dietary cholesterol intake and facilitates the release and clearance of lipoprotein particles. Persistent hypercholesterolemia leads to immune responses against lipoprotein particles that drive atherosclerosis. However, the effect of hypercholesterolemia on hepatic T-cell differentiation remains unknown. OBJECTIVE: To investigate hepatic T-cell subsets upon hypercholesterolemia. METHODS AND RESULTS: We observed that hypercholesterolemia elevated the intrahepatic regulatory T (Treg) cell population and increased the expression of transforming growth factor-ß1 in the liver. Adoptive transfer experiments revealed that intrahepatically differentiated Treg cells relocated to the inflamed aorta in atherosclerosis-prone low-density lipoprotein receptor deficient (Ldlr-/-) mice. Moreover, hypercholesterolemia induced the differentiation of intrahepatic, but not intrasplenic, Th17 cells in wild-type mice, whereas the disrupted liver homeostasis in hypercholesterolemic Ldlr-/- mice led to intrahepatic Th1 cell differentiation and CD11b+CD11c+ leukocyte accumulation. CONCLUSIONS: Our results elucidate a new mechanism that controls intrahepatic T-cell differentiation during atherosclerosis development and indicates that intrahepatically differentiated T cells contribute to the CD4+ T-cell pool in the atherosclerotic aorta.


Asunto(s)
Diferenciación Celular/fisiología , Hipercolesterolemia/sangre , Hipercolesterolemia/patología , Hígado/citología , Hígado/fisiología , Linfocitos T Reguladores/fisiología , Animales , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Distribución Aleatoria
3.
J Autoimmun ; 63: 23-30, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26149776

RESUMEN

The forkhead/winged-helix transcription factor FOXP3 confers suppressive ability to CD4(+)FOXP3(+) regulatory T (Treg) cells. Human Treg cells express several different isoforms of FOXP3 that differ in function. However, the regulation and functional consequences of FOXP3 isoform expression remains poorly understood. In order to study the function of the FOXP3Δ2Δ7 isoform in vivo we generated mice that exclusively expressed a Foxp3 isoform lacking exon 2 and 7. These mice exhibited multi-organ inflammation, increased cytokine production, global T cell activation, activation of antigen-presenting cells and B cell developmental defects, all features that are shared with mice completely deficient in FOXP3. Our results demonstrate that the mouse counterpart of human FOXP3Δ2Δ7 is unable to confer suppressive ability to Treg cells.


Asunto(s)
Factores de Transcripción Forkhead , Linfocitos T Reguladores/metabolismo , Animales , Exones , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Humanos , Activación de Linfocitos/genética , Ratones , Ratones Transgénicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Linfocitos T Reguladores/inmunología
4.
Front Med (Lausanne) ; 6: 76, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31106204

RESUMEN

Activated platelets and mast cells expose the inorganic polymer, polyphosphate (polyP) on their surfaces. PolyP initiates procoagulant and proinflammatory reactions and the polymer has been recognized as a therapeutic target for interference with blood coagulation and vascular hyperpermeability. PolyP content and chain length depend on the specific cell type and energy status, which may affect cellular functions. PolyP metabolism has mainly been studied in bacteria and yeast, but its roles in eukaryotic cells and mammalian systems have remained enigmatic. In this review, we will present an overview of polyP functions, focusing on intra- and extracellular roles of the polymer and discuss open questions that emerge from the current knowledge on polyP regulation.

5.
Front Immunol ; 9: 530, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29593749

RESUMEN

FOXP3 is the lineage-defining transcription factor of CD4+ CD25+ regulatory T cells. While many aspects of its regulation, interaction, and function are conserved among species, alternatively spliced FOXP3 isoforms are expressed only in human cells. This review summarizes current knowledge about alternative splicing of FOXP3 and the specific functions of FOXP3 isoforms in health and disease. Future perspectives in research and the therapeutic potential of manipulating alternative splicing of FOXP3 are discussed.


Asunto(s)
Factores de Transcripción Forkhead/genética , Neoplasias/genética , Empalme Alternativo , Animales , Exones , Humanos , Isoformas de Proteínas/genética
6.
Atherosclerosis ; 267: 27-33, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29100058

RESUMEN

BACKGROUND AND AIMS: The expression of FOXP3 isoforms affects regulatory T (Treg) cell function. Reduced Treg cell function has been associated with coronary artery disease (CAD). However, alternative splicing of FOXP3 in CAD has not been investigated. METHODS: FOXP3 splice variants and IL17A transcripts in peripheral blood mononuclear cells from stable CAD patients and healthy controls were quantified, and FOXP3 isoform expression in response to T cell receptor (TCR) stimulation or LDL was analyzed by flow cytometry. RESULTS: Compared to healthy controls, CAD patients expressed significantly more FOXP3 transcripts that included exon 2, whereas alternative splicing of exon 7 in correlation with IL17A expression was reduced. Moreover, TCR stimulation, as well as exposure to LDL, decreased alternative splicing of FOXP3 in CD4+ T cells in vitro. CONCLUSIONS: Our results demonstrate that blood mononuclear cells in stable CAD patients express a ratio of FOXP3 isoforms that is characteristic for activated CD4+ T cells.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Factores de Transcripción Forkhead/genética , Interleucina-17/metabolismo , Adulto , Anciano , Empalme Alternativo , Linfocitos T CD4-Positivos/citología , Estudios de Casos y Controles , Enfermedad de la Arteria Coronaria/inmunología , Exones , Femenino , Citometría de Flujo , Factores de Transcripción Forkhead/sangre , Factores de Transcripción Forkhead/inmunología , Humanos , Interleucina-17/sangre , Interleucina-17/genética , Leucocitos Mononucleares/metabolismo , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Isoformas de Proteínas , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología
7.
Sci Rep ; 7(1): 15655, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-29142309

RESUMEN

Hypercholesterolemia promotes the inflammation against lipoproteins in atherosclerosis. Development of atherosclerosis is affected by the balance between pro-inflammatory effector T cells and anti-inflammatory regulatory T (Treg) cells. However, phenotype and function of T cell subpopulations in hypercholesterolemia remain to be investigated. Here, we found that cholesterol-containing diet increased the expression of the Treg cell lineage-defining transcription factor FoxP3 among thymocytes and splenocytes. Hypercholesterolemia elevated the FoxP3 expression level and population size of peripheral Treg cells, but did not prevent enhanced proliferation of stimulated T cells. Moreover, cholesterol supplementation in diet as well as in cell culture medium promoted T cell antigen receptor (TCR) signaling in CD4+ T cells. Our results demonstrate that hypercholesterolemia enhances TCR stimulation, Treg cell development as well as T cell proliferation. Thus, our findings may help to understand why hypercholesterolemia correlates with altered CD4+ T cell responses.


Asunto(s)
Aterosclerosis/inmunología , Factores de Transcripción Forkhead/genética , Hipercolesterolemia/inmunología , Inflamación/inmunología , Receptores de Antígenos de Linfocitos T/genética , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linaje de la Célula/inmunología , Colesterol en la Dieta/farmacología , Factores de Transcripción Forkhead/inmunología , Humanos , Hipercolesterolemia/genética , Hipercolesterolemia/patología , Inflamación/genética , Inflamación/patología , Lipoproteínas/metabolismo , Ratones Noqueados , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de LDL/genética , Transducción de Señal/inmunología , Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Timocitos/metabolismo , Timocitos/patología
8.
Sci Rep ; 5: 14674, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26441347

RESUMEN

CD4(+)FOXP3(+) regulatory T (Treg) cells are essential for maintaining immunological self-tolerance. Treg cell development and function depend on the transcription factor FOXP3, which is present in several distinct isoforms due to alternative splicing. Despite the importance of FOXP3 in the proper maintenance of Treg cells, the regulation and functional consequences of FOXP3 isoform expression remains poorly understood. Here, we show that in human Treg cells IL-1ß promotes excision of FOXP3 exon 7. FOXP3 is not only expressed by Treg cells but is also transiently expressed when naïve T cells differentiate into Th17 cells. Forced splicing of FOXP3 into FOXP3Δ2Δ7 strongly favored Th17 differentiation in vitro. We also found that patients with Crohn's disease express increased levels of FOXP3 transcripts lacking exon 7, which correlate with disease severity and IL-17 production. Our results demonstrate that alternative splicing of FOXP3 modulates T cell differentiation. These results highlight the importance of characterizing FOXP3 expression on an isoform basis and suggest that immune responses may be manipulated by modulating the expression of FOXP3 isoforms, which has broad implications for the treatment of autoimmune diseases.


Asunto(s)
Empalme Alternativo , Diferenciación Celular , Enfermedad de Crohn/genética , Factores de Transcripción Forkhead/genética , Interleucina-17/metabolismo , Interleucina-1beta/farmacología , Células Th17/citología , Western Blotting , Células Cultivadas , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/patología , Humanos , Tolerancia Inmunológica/inmunología , Activación de Linfocitos , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Células Th17/metabolismo
9.
PLoS One ; 4(7): e6104, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19568423

RESUMEN

BACKGROUND: Phenotype and function of regulatory T cells (Treg) largely depend on the presence of the transcription factor FOXP3. In contrast to mice, human Treg cells express isoforms of this protein. Besides the full length version (FOXP3fl), an isoform lacking the exon 2 (FOXP3Delta2) is co-expressed in comparable amounts. Recently, a third splice variant has been described that in addition to exon 2 also misses exon 7 (FOXP3Delta2Delta7). Exon 7 encodes for a leucine zipper motif commonly used as structural dimerization element. Mutations in exon 7 have been linked to IPEX, a severe autoimmune disease suggested to be caused by impaired dimerization of the FOXP3 protein. PRINCIPAL FINDINGS: This study shows that the lack of exon 7 does not affect (homo-) dimerization. Moreover, the interaction of FOXP3Delta2Delta7 to RUNX1, NFAT and NF-kB appeared to be unchanged in co-immunoprecipitation experiments and reporter gene assays, when compared to FOXP3fl and FOXP3Delta2. Nevertheless, retroviral transduction with FOXP3Delta2Delta7 failed to induce the typical Treg-associated phenotype. The expression of FOXP3-induced surface molecules such as CD25 and CTLA-4 were not enhanced in FOXP3Delta2Delta7 transduced CD4+ T cells, which also failed to exhibit any suppressive capacity. Notably, however, co-expression of FOXP3fl with FOXP3Delta2Delta7 resulted in a reduction of CD25 expression by a dominant negative effect. CONCLUSIONS: The leucine zipper of FOXP3 does not mediate dimerization or interaction with NFAT, NF-kB and RUNX1, but is indispensable for the characteristic phenotype and function in Treg cells. FOXP3Delta2Delta7 could play a role in regulating the function of the other FOXP3 isoforms and may be involved in cancer pathogenesis, as it is overexpressed by certain malignant cells.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , Leucina Zippers , Animales , Secuencia de Bases , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Cartilla de ADN , Dimerización , Exones , Factores de Transcripción Forkhead/química , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , FN-kappa B/fisiología , Factores de Transcripción NFATC/fisiología , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fracciones Subcelulares/metabolismo , Linfocitos T Reguladores/inmunología , Transcripción Genética/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA