Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 209(10): 1906-1917, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36426957

RESUMEN

Rheumatoid arthritis (RA) is characterized by autoimmune joint destruction with debilitating consequences. Despite treatment advancements with biologic therapies, a significant proportion of RA patients show an inadequate clinical response, and restoration of immune self-tolerance represents an unmet therapeutic need. We have previously described a tolerogenic phenotype of plasmacytoid dendritic cells (pDCs) in RA patients responding to anti-TNF-α agents. However, the molecular mechanisms involved in tolerogenic reprogramming of pDCs in RA remain elusive. In this study, guided by transcriptomic analysis of CD303+CD123+ pDCs from RA patients in remission, we revealed enhanced expression of IL-6R and its downstream signaling compared with healthy pDCs. Functional assessment demonstrated that IL-6R engagement resulted in marked reduction of TNF-α secretion by pDCs whereas intracellular TNF-α was significantly increased. Accordingly, pharmacologic inhibition of IL-6R signaling restored TNF-α secretion levels by pDCs. Mechanistic analysis demonstrated impaired activity and decreased lysosomal degradation of ADAM17 (a disintegrin and metalloproteinase 17) sheddase in pDCs, which is essential for TNF-α cleavage. Importantly, reduction of TNF-α secretion by IL-6-treated pDCs attenuated the inflammatory potential of RA patient-derived synovial fibroblasts. Collectively, these findings position pDCs as an important source of TNF-α in RA pathogenesis and unravel an anti-inflammatory mechanism of IL-6 by limiting the pDC-derived TNF-α secretion.


Asunto(s)
Artritis Reumatoide , Interleucina-6 , Humanos , Inhibidores del Factor de Necrosis Tumoral , Células Dendríticas , Transducción de Señal , Factor de Necrosis Tumoral alfa
2.
Eur J Immunol ; 51(1): 125-137, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33107588

RESUMEN

Calcitriol and 9-cis retinoic acid (9cRA) play a fundamental role in shaping the adaptive immune response by altering the Ig profile and the differentiation of B cells, controlled by their corresponding nuclear receptors, VDR and RAR. Herein, after the establishment of a plasmablast differentiation culture, we investigated how both ligands modulate human naïve B cell differentiation and to which extent VDR/RXR and RAR/RXR signaling interferes. Calcitriol and 9cRA mediated activation of purified naïve B cells resulted in a strong differentiation of CD27+ CD38+ plasmablasts and antibody secretion. The significant IgA response was preceded by a strong induction of α-germline transcription (GLT). Induction of αGLT and consecutively IgA secretion driven by calcitriol is a novel observation and we show by magnetic chromatin IP that this was mediated by recruitment of the VDR to the TGF-ß promoter thus inducing TGF-ß expression. Finally, as revealed by transcriptomic profiling calcitriol and 9cRA modulate several signals required for differentiation and isotype switching in a noncompeting but rather additive manner. Calcitriol and 9cRA participate in the control of the IgA response in human activated naïve B cells. The balance between both ligands may be an important factor in channeling humoral immune responses toward a protective direction.


Asunto(s)
Alitretinoína/inmunología , Alitretinoína/farmacología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Calcitriol/inmunología , Calcitriol/farmacología , Inmunoglobulina A/biosíntesis , Inmunidad Adaptativa/efectos de los fármacos , Linfocitos B/citología , Sitios de Unión/genética , Ligando de CD40/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Células Cultivadas , Proteínas de Unión al GTP/genética , Expresión Génica , Humanos , Cambio de Clase de Inmunoglobulina/efectos de los fármacos , Cambio de Clase de Inmunoglobulina/inmunología , Interleucina-4/inmunología , Ligandos , Activación de Linfocitos , Células Plasmáticas/citología , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/inmunología , Regiones Promotoras Genéticas , Proteína Glutamina Gamma Glutamiltransferasa 2 , Receptores de Calcitriol/inmunología , Receptores de Ácido Retinoico/inmunología , Receptores X Retinoide/inmunología , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/genética , Transglutaminasas/genética , Vitamina D3 24-Hidroxilasa/genética
3.
Nat Immunol ; 11(11): 1057-62, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20935646

RESUMEN

After being activated by antigen, helper T lymphocytes switch from a resting state to clonal expansion. This switch requires inactivation of the transcription factor Foxo1, a suppressor of proliferation expressed in resting helper T lymphocytes. In the early antigen-dependent phase of expansion, Foxo1 is inactivated by antigen receptor-mediated post-translational modifications. Here we show that in the late phase of expansion, Foxo1 was no longer post-translationally regulated but was inhibited post-transcriptionally by the interleukin 2 (IL-2)-induced microRNA miR-182. Specific inhibition of miR-182 in helper T lymphocytes limited their population expansion in vitro and in vivo. Our results demonstrate a central role for miR-182 in the physiological regulation of IL-2-driven helper T cell-mediated immune responses and open new therapeutic possibilities.


Asunto(s)
Interleucina-2/inmunología , MicroARNs/inmunología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Artritis/inmunología , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
4.
Nature ; 507(7492): 366-370, 2014 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-24572363

RESUMEN

B lymphocytes have critical roles as positive and negative regulators of immunity. Their inhibitory function has been associated primarily with interleukin 10 (IL-10) because B-cell-derived IL-10 can protect against autoimmune disease and increase susceptibility to pathogens. Here we identify IL-35-producing B cells as key players in the negative regulation of immunity. Mice in which only B cells did not express IL-35 lost their ability to recover from the T-cell-mediated demyelinating autoimmune disease experimental autoimmune encephalomyelitis (EAE). In contrast, these mice displayed a markedly improved resistance to infection with the intracellular bacterial pathogen Salmonella enterica serovar Typhimurium as shown by their superior containment of the bacterial growth and their prolonged survival after primary infection, and upon secondary challenge, compared to control mice. The increased immunity found in mice lacking IL-35 production by B cells was associated with a higher activation of macrophages and inflammatory T cells, as well as an increased function of B cells as antigen-presenting cells (APCs). During Salmonella infection, IL-35- and IL-10-producing B cells corresponded to two largely distinct sets of surface-IgM(+)CD138(hi)TACI(+)CXCR4(+)CD1d(int)Tim1(int) plasma cells expressing the transcription factor Blimp1 (also known as Prdm1). During EAE, CD138(+) plasma cells were also the main source of B-cell-derived IL-35 and IL-10. Collectively, our data show the importance of IL-35-producing B cells in regulation of immunity and highlight IL-35 production by B cells as a potential therapeutic target for autoimmune and infectious diseases. This study reveals the central role of activated B cells, particularly plasma cells, and their production of cytokines in the regulation of immune responses in health and disease.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Inmunidad/inmunología , Interleucinas/metabolismo , Infecciones por Salmonella/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos CD40/inmunología , Femenino , Humanos , Interleucina-10/metabolismo , Interleucinas/inmunología , Activación de Linfocitos , Macrófagos/citología , Macrófagos/inmunología , Masculino , Ratones , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Infecciones por Salmonella/microbiología , Linfocitos T/inmunología , Receptor Toll-Like 4/inmunología
5.
Ann Rheum Dis ; 77(2): 300-308, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29191820

RESUMEN

OBJECTIVE: Rheumatoid arthritis (RA) accompanies infiltration and activation of monocytes in inflamed joints. We investigated dominant alterations of RA monocytes in bone marrow (BM), blood and inflamed joints. METHODS: CD14+ cells from BM and peripheral blood (PB) of patients with RA and osteoarthritis (OA) were profiled with GeneChip microarrays. Detailed functional analysis was performed with reference transcriptomes of BM precursors, monocyte blood subsets, monocyte activation and mobilisation. Cytometric profiling determined monocyte subsets of CD14++CD16-, CD14++CD16+ and CD14+CD16+ cells in BM, PB and synovial fluid (SF) and ELISAs quantified the release of activation markers into SF and serum. RESULTS: Investigation of genes differentially expressed between RA and OA monocytes with reference transcriptomes revealed gene patterns of early myeloid precursors in RA-BM and late myeloid precursors along with reduced terminal differentiation to CD14+CD16+monocytes in RA-PB. Patterns associated with tumor necrosis factor/lipopolysaccharide (TNF/LPS) stimulation were weak and more pronounced in RA-PB than RA-BM. Cytometric phenotyping of cells in BM, blood and SF disclosed differences related to monocyte subsets and confirmed the reduced frequency of terminally differentiated CD14+CD16+monocytes in RA-PB. Monocyte activation in SF was characterised by the predominance of CD14++CD16++CD163+HLA-DR+ cells and elevated concentrations of sCD14, sCD163 and S100P. CONCLUSION: Patterns of less mature and less differentiated RA-BM and RA-PB monocytes suggest increased turnover with accelerated monocytopoiesis, BM egress and migration into inflamed joints. Predominant activation in the joint indicates the action of local and primary stimuli, which may also promote adaptive immune triggering through monocytes, potentially leading to new diagnostic and therapeutic strategies.


Asunto(s)
Artritis Reumatoide/patología , Médula Ósea/patología , Articulaciones/patología , Monocitos/citología , Artritis Reumatoide/genética , Artritis Reumatoide/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Perfilación de la Expresión Génica/métodos , Humanos , Monocitos/metabolismo , Monocitos/patología , Osteoartritis/genética , Osteoartritis/inmunología , Osteoartritis/patología , Líquido Sinovial/citología
6.
Blood ; 128(26): 3125-3136, 2016 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-27702798

RESUMEN

Metabolic changes drive monocyte differentiation and fate. Although abnormal mitochondria metabolism and innate immune responses participate in the pathogenesis of many inflammatory disorders, molecular events regulating mitochondrial activity to control life and death in monocytes remain poorly understood. We show here that, in human monocytes, microRNA-125b (miR-125b) attenuates the mitochondrial respiration through the silencing of the BH3-only proapoptotic protein BIK and promotes the elongation of the mitochondrial network through the targeting of the mitochondrial fission process 1 protein MTP18, leading to apoptosis. Proinflammatory activation of monocyte-derived macrophages is associated with a concomitant increase in miR-125b expression and decrease in BIK and MTP18 expression, which lead to reduced oxidative phosphorylation and enhanced mitochondrial fusion. In a chronic inflammatory systemic disorder, CD14+ blood monocytes display reduced miR-125b expression as compared with healthy controls, inversely correlated with BIK and MTP18 messenger RNA expression. Our findings not only identify BIK and MTP18 as novel targets for miR-125b that control mitochondrial metabolism and dynamics, respectively, but also reveal a novel function for miR-125b in regulating metabolic adaptation of monocytes to inflammation. Together, these data unravel new molecular mechanisms for a proapoptotic role of miR-125b in monocytes and identify potential targets for interfering with excessive inflammatory activation of monocytes in inflammatory disorders.


Asunto(s)
Inflamación/genética , Inflamación/patología , MicroARNs/metabolismo , Mitocondrias/metabolismo , Dinámicas Mitocondriales/genética , Monocitos/metabolismo , Monocitos/patología , Anciano , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Polaridad Celular/genética , Respiración de la Célula/genética , Femenino , Regulación de la Expresión Génica , Silenciador del Gen , Células HEK293 , Humanos , Receptores de Lipopolisacáridos/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , MicroARNs/genética , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Modelos Biológicos , Receptor Toll-Like 4/metabolismo
7.
Immunity ; 30(5): 721-30, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19427242

RESUMEN

CD4(+) T lymphocytes are key to immunological memory. Here we show that in the memory phase of specific immune responses, most of the memory CD4(+) T lymphocytes had relocated into the bone marrow (BM) within 3-8 weeks after their generation-a process involving integrin alpha2. Antigen-specific memory CD4(+) T lymphocytes highly expressed Ly-6C, unlike most splenic CD44(hi)CD62L(-) CD4(+) T lymphocytes. In adult mice, more than 80% of Ly-6C(hi)CD44(hi)CD62L(-) memory CD4(+) T lymphocytes were in the BM. In the BM, they associated to IL-7-expressing VCAM-1(+) stroma cells. Gene expression and proliferation were downregulated, indicating a resting state. Upon challenge with antigen, they rapidly expressed cytokines and CD154 and efficiently induced the production of high-affinity antibodies by B lymphocytes. Thus, in the memory phase of immunity, memory helper T cells are maintained in BM as resting but highly reactive cells in survival niches defined by IL-7-expressing stroma cells.


Asunto(s)
Médula Ósea/inmunología , Linfocitos T CD4-Positivos/inmunología , Memoria Inmunológica , Animales , Antígenos Ly/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/metabolismo , Regulación hacia Abajo/inmunología , Expresión Génica , Integrina alfa2/inmunología , Interleucina-7/inmunología , Interleucina-7/metabolismo , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Células del Estroma/inmunología , Células del Estroma/metabolismo
8.
J Cell Physiol ; 232(6): 1326-1336, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27636101

RESUMEN

Familial Mediterranean fever (FMF) is an autosomal recessive disease characterized by recurrent, acute, and self-limiting attacks of fever. Mutations in MEFV gene encoding pyrin account for FMF, but the high number of heterozygote patients with typical symptoms of the disease has driven a number of alternative aetiopathogenic hypotheses. The MEFV gene was knocked down in human myelomonocytic cells that express endogenous pyrin to identify deregulated microRNAs (miRNAs). Microarray analyses revealed 29 significantly differentially expressed miRNAs implicated in pathways associated with cellular integrity and survival. Implementation of in silico gene network prediction algorithms and bioinformatics analyses showed that miR-4520a is predicted to target genes implicated in autophagy through regulation of RHEB/mTOR signaling. Differential expression levels of RHEB were confirmed by luciferase reporter gene assays providing further evidence that is directly targeted by miR-4520a. Although the relative expression levels of miR-4520a were variable among FMF patients, the statistical expression of miR-4520a was different between FMF mutation carriers and controls (P = 0.0061), indicating an association between miR-4520a expression and MEFV mutations. Comparison between FMF patients bearing the M694V mutation, associated with severe disease, and healthy controls showed a significant increase in miR-4520a expression levels (P = 0.00545). These data suggest that RHEB, the main activator of mTOR signaling, is a valid target of miR-4520a with the relative expression levels of the latter being significantly deregulated in FMF patients and highly dependent on the presence of pyrin mutations, especially of the M694V type. These results suggest a role of deregulated autophagy in the pathogenesis of FMF. J. Cell. Physiol. 232: 1326-1336, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Fiebre Mediterránea Familiar/genética , Perfilación de la Expresión Génica , MicroARNs/genética , Mutación/genética , Pirina/genética , Adulto , Estudios de Casos y Controles , Línea Celular , Femenino , Redes Reguladoras de Genes , Humanos , Luciferasas/metabolismo , Masculino , MicroARNs/metabolismo , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro
9.
Blood ; 125(11): 1739-48, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25573986

RESUMEN

Specific serum antibodies mediating humoral immunity and autoimmunity are provided by mature plasma cells (PC) residing in the bone marrow (BM), yet their dynamics and composition are largely unclear. We here characterize distinct subsets of human PC differing by CD19 expression. Unlike CD19(+) PC, CD19(-) PC were restricted to BM, expressed predominantly IgG, and they carried a prosurvival, distinctly mature phenotype, that is, HLA-DR(low)Ki-67(-)CD95(low)CD28(+)CD56(+/-), with increased BCL2 and they resisted their mobilization from the BM after systemic vaccination. Fewer mutations within immunoglobulin VH rearrangements of CD19(-) BMPC may indicate their differentiation in early life. Their resistance to in vivo B-cell depletion, that is, their independency from supply with new plasmablasts, is consistent with long-term stability of this PC subset in the BM. Moreover, CD19(-) PC were detectable in chronically inflamed tissues and secreted autoantibodies. We propose a multilayer model of PC memory in which CD19(+) and CD19(-) PC represent dynamic and static components, respectively, permitting both adaptation and stability of humoral immune protection.


Asunto(s)
Antígenos CD19/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Inmunoglobulina G/metabolismo , Células Plasmáticas/inmunología , Suero Antilinfocítico/administración & dosificación , Células de la Médula Ósea/clasificación , Ácidos Borónicos/administración & dosificación , Bortezomib , Diferenciación Celular , Supervivencia Celular , Vacuna contra Difteria y Tétanos/administración & dosificación , Humanos , Inmunidad Humoral , Memoria Inmunológica , Inflamación/inmunología , Inflamación/patología , Depleción Linfocítica , Modelos Inmunológicos , Mutación , Fenotipo , Células Plasmáticas/clasificación , Células Plasmáticas/citología , Pirazinas/administración & dosificación , Recombinación V(D)J
10.
J Immunol ; 194(12): 5812-24, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25948818

RESUMEN

Foxp3(+) regulatory T cell (Treg)-based immunotherapy holds promise for autoimmune diseases. However, this effort has been hampered by major caveats, including the low frequency of autoantigen-specific Foxp3(+) Tregs and lack of understanding of their molecular and cellular targets, in an unmanipulated wild-type (WT) immune repertoire. In this study, we demonstrate that infusion of myelin in WT mice results in the de novo induction of myelin-specific Foxp3(+) Tregs in WT mice and amelioration of experimental autoimmune encephalomyelitis. Myelin-specific Foxp3(+) Tregs exerted their effect both by diminishing Ag-bearing inflammatory dendritic cell (iDC) recruitment to lymph nodes and by impairing their function. Transcriptome analysis of ex vivo-isolated Treg-exposed iDCs showed significant enrichment of transcripts involved in functional properties of iDCs, including chemotaxis-related genes. To this end, CCR7 expression by iDCs was significantly downregulated in tolerant mice and this was tightly regulated by the presence of IL-10. Collectively, our data demonstrate a novel model for deciphering the Ag-specific Foxp3(+) Treg-mediated mechanisms of tolerance and delineate iDCs as a Foxp3(+) Treg cellular target in unmanipulated mice.


Asunto(s)
Autoantígenos/inmunología , Células Dendríticas/inmunología , Inflamación/inmunología , Ganglios Linfáticos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Autoinmunidad , Quimiotaxis/inmunología , Análisis por Conglomerados , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Tolerancia Inmunológica , Inflamación/genética , Inflamación/metabolismo , Interleucina-10/metabolismo , Depleción Linfocítica , Ratones , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Glicoproteína Mielina-Oligodendrócito/inmunología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Receptores CCR7/genética , Receptores CCR7/metabolismo , Transducción de Señal , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T Reguladores/metabolismo
11.
Proc Natl Acad Sci U S A ; 111(25): 9229-34, 2014 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-24927527

RESUMEN

In the bone marrow, a population of memory T cells has been described that promotes efficient secondary immune responses and has been considered to be preactivated, owing to its expression of CD69 and CD25. Here we show that human bone marrow professional memory T cells are not activated but are resting in terms of proliferation, transcription, and mobility. They are in the G0 phase of the cell cycle, and their transcriptome is that of resting T cells. The repertoire of CD4(+) bone marrow memory T cells compared with CD4(+) memory T cells from the blood is significantly enriched for T cells specific for cytomegalovirus-pp65 (immunodominant protein), tetanus toxoid, measles, mumps, and rubella. It is not enriched for vaccinia virus and Candida albicans-MP65 (immunodominant protein), typical pathogens of skin and/or mucosa. CD4(+) memory T cells specific for measles are maintained nearly exclusively in the bone marrow. Thus, CD4(+) memory T cells from the bone marrow provide long-term memory for systemic pathogens.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/inmunología , Células de la Médula Ósea/inmunología , Linfocitos T CD4-Positivos/inmunología , Memoria Inmunológica/fisiología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Lectinas Tipo C/inmunología , Fase de Descanso del Ciclo Celular/inmunología , Adulto , Células de la Médula Ósea/citología , Linfocitos T CD4-Positivos/citología , Femenino , Humanos , Masculino , Persona de Mediana Edad
12.
Eur J Immunol ; 45(4): 975-87, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25639669

RESUMEN

It is believed that memory CD8(+) T cells are maintained in secondary lymphoid tissues, peripheral tissues, and BM by homeostatic proliferation. Their survival has been shown to be dependent on IL-7, but it is unclear where they acquire it. Here we show that in murine BM, memory CD8(+) T cells individually colocalize with IL-7(+) reticular stromal cells. The T cells are resting in terms of global transcription and do not express markers of activation, for example, 4-1BB (CD137), IL-2, or IFN-γ, despite the expression of CD69 on about 30% of the cells. Ninety-five percent of the memory CD8(+) T cells in BM are in G0 phase of cell cycle and do not express Ki-67. Less than 1% is in S/M/G2 of cell cycle, according to propidium iodide staining. While previous publications have estimated the extent of proliferation of CD8(+) memory T cells on the basis of BrdU incorporation, we show here that BrdU itself induces proliferation of CD8(+) memory T cells. Taken together, the present results suggest that CD8(+) memory T cells are maintained as resting cells in the BM in dedicated niches with their survival conditional on IL-7 receptor signaling.


Asunto(s)
Células de la Médula Ósea/citología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Fase de Descanso del Ciclo Celular/inmunología , Células del Estroma/inmunología , Animales , Antígenos CD/biosíntesis , Antígenos de Diferenciación de Linfocitos T/biosíntesis , Células de la Médula Ósea/inmunología , Proliferación Celular , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Interleucina-7/inmunología , Antígeno Ki-67/biosíntesis , Lectinas Tipo C/biosíntesis , Ratones , Ratones Endogámicos C57BL , Transcripción Genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis
13.
Eur J Immunol ; 45(4): 1192-205, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25486906

RESUMEN

Repeatedly activated T helper 1 (Th1) cells present during chronic inflammation can efficiently adapt to the inflammatory milieu, for example, by expressing the transcription factor Twist1, which limits the immunopathology caused by Th1 cells. Here, we show that in repeatedly activated murine Th1 cells, Twist1 and T-bet induce expression of microRNA-148a (miR-148a). miR-148a regulates expression of the proapoptotic gene Bim, resulting in a decreased Bim/Bcl2 ratio. Inhibition of miR-148a by antagomirs in repeatedly activated Th1 cells increases the expression of Bim, leading to enhanced apoptosis. Knockdown of Bim expression by siRNA in miR-148a antagomir-treated cells restores viability of the Th1 cells, demonstrating that miR-148a controls survival by regulating Bim expression. Thus, Twist1 and T-bet not only control the differentiation and function of Th1 cells, but also their persistence in chronic inflammation.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Apoptosis/genética , Regulación de la Expresión Génica , Proteínas de la Membrana/genética , MicroARNs/fisiología , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas de Dominio T Box/fisiología , Células TH1/inmunología , Proteína 1 Relacionada con Twist/metabolismo , Animales , Artritis Reumatoide/inmunología , Proteína 11 Similar a Bcl2 , Supervivencia Celular/inmunología , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Proteínas Nucleares/genética , Interferencia de ARN , ARN Interferente Pequeño , Proteínas de Dominio T Box/genética , Proteína 1 Relacionada con Twist/genética
14.
Ann Rheum Dis ; 75(2): 413-21, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25414238

RESUMEN

OBJECTIVE: Arthritis is a chronic inflammatory disease characterised by immune cell infiltration and mesenchymal cell expansion in the joints. Although the role of immune cells in arthritis is well characterised, the development of mesenchymal cell hyperplasia needs to be better defined. Here, we analysed the role of the ribosomal S6 kinase Rsk2, which we found to be highly activated in joints of patients with arthritis, in the development of mesenchymal cell hyperplasia. METHODS: We genetically inactivated Rsk2 in the tumour necrosis factor (TNF)-α transgenic (TNFtg) mice, an animal model for human inflammatory arthritis. Clinical and histological signs of arthritis as well as molecular markers of inflammation and joint destruction were quantified. Fibroblast-like synoviocytes (FLS) were characterised in vitro and the effect of Rsk2 deletion on the pattern of gene expression was determined. RESULTS: Rsk2 deficiency in TNFtg mice results in earlier and exacerbated inflammation as well as increased bone and cartilage destruction. The production of inflammatory cytokines, matrix metalloproteinases and osteoclastogenic molecules was significantly increased in vivo upon Rsk2 inactivation. Bone marrow deficient in Rsk2 could not transfer this phenotype, indicating that Rsk2 expression in mesenchymal cells controls the course of arthritis. Indeed, Rsk2 deficiency was associated with a more activated phenotype and higher proliferative capacity of FLS, thereby increasing cytokines and production of matrix proteinases. CONCLUSIONS: Rsk2 emerges as a key regulator of mesenchymal cell numbers in the joint and thereby could be targeted to control the inflammatory and tissue-destructive feature of joints in arthritis.


Asunto(s)
Artritis Experimental/patología , Fibroblastos/patología , Proteínas Quinasas S6 Ribosómicas 90-kDa/fisiología , Membrana Sinovial/patología , Animales , Artritis Experimental/metabolismo , Proliferación Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Hiperplasia/genética , Hiperplasia/metabolismo , Inflamación/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Transgénicos , Proteínas Quinasas S6 Ribosómicas 90-kDa/deficiencia , Membrana Sinovial/metabolismo , Factor de Necrosis Tumoral alfa/genética
15.
Eur J Immunol ; 44(6): 1615-21, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24643722

RESUMEN

B-cell-derived interleukin-10 (IL-10) is known to act in a paracrine fashion to suppress inflammation. Here, we show that IL-10 also acts in an autocrine manner to regulate the differentiation of activated human B cells. We report that IL-10 expression is not restricted to a dedicated B-cell subset, but is induced transiently in peripheral human naïve, memory, and CD5(+) B cells alike upon activation. Global transcriptome comparison of activated human B cells, secreting IL-10 or not, identified 138 differentially regulated genes, most of which were associated with differentiation into antibody-secreting cells and reflecting autocrine IL-10 signaling. We monitored the differentiation of IL-10-secreting B cells and determined the effect of IL-10-blocking antibodies against its autocrine and paracrine signaling. IL-10 signaling promoted the differentiation of activated IL-10-secreting B cells into IgM- or IgG-secreting cells, but was dispensable for IgA secretion. Our data imply that B-cell-derived IL-10 not only suppresses immune reactions via paracrine mechanisms, but can also contribute to the differentiation of IL-10-secreting B cells into IgM- and IgG-secreting plasmablasts through both autocrine and paracrine signaling.


Asunto(s)
Comunicación Autocrina/inmunología , Diferenciación Celular/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Interleucina-10/inmunología , Comunicación Paracrina/inmunología , Células Plasmáticas/inmunología , Transducción de Señal/fisiología , Femenino , Humanos , Inmunoglobulina G/metabolismo , Inmunoglobulina M/metabolismo , Memoria Inmunológica/fisiología , Interleucina-10/metabolismo , Masculino , Células Plasmáticas/citología , Células Plasmáticas/metabolismo
16.
J Hepatol ; 60(6): 1143-50, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24560659

RESUMEN

BACKGROUND & AIMS: The enterohepatic circuit of T cells may be responsible for the development of autoimmune liver disease. We employed transgenic mice to characterize phenotype and migration patterns of CD8 T cells activated in liver and gut. METHODS: We studied the migration of antigen-specific CD8 T cells primed in liver or gut after transfer in wild-type mice or mice that express ovalbumin in liver or gut. We performed transcriptome analysis of these two distinct T cell populations and confirmed our findings by flow cytometry. RESULTS: Specific migration patterns were induced by activation of CD8 T cells in gut or liver. Gut-activated CD8 T cells expressed α4ß7 and CCR9 and migrated to the gut and to the liver. Liver-activated T cells expressed integrins α4, α6, ß1, α4ß7 as well as CD62L, Ly6C, and neuropilin-1 and retained the capability to re-circulate through lymph nodes. Presence of the antigen increased retention of both types of activated T cells in the liver, but migration of liver-activated T cells to the gut was prohibited. CONCLUSIONS: CD8 T cells primed in the liver in vivo are not capable of migrating to the gut, implying that the enterohepatic circuit of CD8 T cells is in fact a one-way road from the gut to the liver. Priming of CD8 T cells in the liver results in a distinct phenotype with attributes of central memory cells and induces a unique homing pattern. Gut-primed T cells preferentially home to the liver, in principle enabling them to induce autoimmune liver disease.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Movimiento Celular/inmunología , Colangitis Esclerosante/inmunología , Circulación Enterohepática/inmunología , Hepatitis Autoinmune/inmunología , Animales , Linfocitos T CD8-positivos/citología , Colangitis Esclerosante/genética , Modelos Animales de Enfermedad , Hepatitis Autoinmune/genética , Intestinos/inmunología , Hígado/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovalbúmina/genética , Ovalbúmina/inmunología , Fenotipo , Transcriptoma/inmunología
17.
Eur J Immunol ; 43(9): 2497-506, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23716169

RESUMEN

Pluripotent hematopoietic stem cells and multipotent myeloid/lymphoid progenitors express miR-221 and miR-222. When Pax5 expression commits these progenitors to monopotent pre-B lymphocytes the two microRNAs (miRNAs) are downregulated. Upon transplantation, stem cells and progenitors can reside in the BM, while pre-B cells, after their commitment, no longer do so. Retrovirally transduced, doxycycline-induced overexpression of either miR-221 or miR-222 in pre-B-I cells does not revert their monopotency to multipotency. However, upon transplantation miR-221, but not miR-222, transduced pre-B-I cells regain the capacity to reside in the BM. Upon subsequent termination of miR-221-expression by removal of doxycycline, the transplanted cells leave the BM again. Microarray analyses identified 25 downregulated miR-221-target genes, which could function to localize phases of B-lymphocyte development in BM before and after commitment.


Asunto(s)
MicroARNs/metabolismo , Factor de Transcripción PAX5/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Células Precursoras de Linfocitos B/trasplante , Animales , Antígenos CD19/biosíntesis , Linfocitos B/inmunología , Linfocitos B/trasplante , Médula Ósea/inmunología , Células de la Médula Ósea/citología , Diferenciación Celular , Línea Celular , Movimiento Celular/inmunología , Regulación hacia Abajo , Doxiciclina , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/genética , Células Progenitoras Linfoides/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Progenitoras Mieloides/metabolismo , Factor de Transcripción PAX5/genética , Células Madre Pluripotentes/metabolismo
18.
Eur J Immunol ; 40(8): 2165-73, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20518031

RESUMEN

Organization of the stromal compartments in secondary lymphoid tissue is a prerequisite for an efficient immune reaction. In particular, follicular dendritic cells (FDC) are pivotal for the activation and differentiation of B cells. To investigate the development of FDC, FDC together with tightly associated B cells (FDC networks) were micro-dissected from frozen tissue sections and follicular B cells were sorted by FACS. Using an in silico subtraction approach, gene expression of FDC was determined and compared with that of follicular stromal cells micro-dissected from the spleen of SCID mice. Nearly 90% of the FDC genes were expressed in follicular stromal cells of the SCID mouse, providing further evidence that FDC develop from the residual network of reticular cells. Thus, it suggests that rather minor modifications in the gene expression profile are sufficient for differentiation into mature FDC. The analysis of different immune-deficient mouse strains shows that a complex pattern of gene regulation controls the development of residual stromal cells into mature FDC. The in silico subtraction approach provides a molecular framework within which to determine the diverse roles of FDC in support of B cells and to investigate the differentiation of FDC from their mesenchymal precursor cells.


Asunto(s)
Células Dendríticas Foliculares/metabolismo , Células Madre Mesenquimatosas/metabolismo , Bazo/patología , Células del Estroma/metabolismo , ADP-Ribosil Ciclasa/biosíntesis , Animales , Antígenos CD/biosíntesis , Antígenos de Diferenciación/biosíntesis , Linfocitos B/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Separación Celular , Células Cultivadas , Células Dendríticas Foliculares/inmunología , Células Dendríticas Foliculares/patología , Citometría de Flujo , Proteínas Ligadas a GPI/biosíntesis , Perfilación de la Expresión Génica , Activación de Linfocitos/genética , Células Madre Mesenquimatosas/patología , Ratones , Ratones SCID , Bazo/metabolismo , Células del Estroma/patología
19.
Eur J Immunol ; 40(11): 2993-3006, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21061432

RESUMEN

Th1 cells are prominent in inflamed tissue, survive conventional immunosuppression, and are believed to play a pivotal role in driving chronic inflammation. Here, we identify homeobox only protein (Hopx) as a critical and selective regulator of the survival of Th1 effector/memory cells, both in vitro and in vivo. Expression of Hopx is induced by T-bet and increases upon repeated antigenic restimulation of Th1 cells. Accordingly, the expression of Hopx is low in peripheral, naïve Th cells, but highly up-regulated in terminally differentiated effector/memory Th1 cells of healthy human donors. In murine Th1 cells, Hopx regulates the expression of genes involved in regulation of apoptosis and survival and makes them refractory to Fas-induced apoptosis. In vivo, adoptively transferred Hopx-deficient murine Th1 cells do not persist. Consequently, they cannot induce chronic inflammation in murine models of transfer-induced colitis and arthritis, demonstrating a key role of Hopx for Th1-mediated immunopathology.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Proteínas de Homeodominio/inmunología , Memoria Inmunológica , Células TH1/inmunología , Proteínas Supresoras de Tumor/inmunología , Animales , Apoptosis/inmunología , Artritis/inmunología , Artritis/patología , Supervivencia Celular/inmunología , Colitis/inmunología , Colitis/patología , Modelos Animales de Enfermedad , Humanos , Inflamación/inmunología , Inflamación/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptor fas/inmunología
20.
Arthritis Res Ther ; 23(1): 246, 2021 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-34560894

RESUMEN

BACKGROUND: In patients with axial spondyloarthritis (axSpA), monocytes show a pre-activated phenotype. Gut inflammation is a trigger of monocyte activation and may also affect their development in the bone marrow (BM). As gut inflammation is commonly observed in axSpA patients, we performed a detailed analysis of monocyte transcriptomes of axSpA patients in two cohorts and searched for signs of activation and developmental adaptations as putative imprints of gut inflammation. METHODS: Transcriptomes of blood CD14+ monocytes of HLA-B27+ axSpA patients and healthy controls (HC) were generated by microarrays from cohort 1 and by RNA-sequencing from cohort 2. Differentially expressed genes from both analyses were subjected to gene set enrichment analysis (GSEA) and to co-expression analysis in reference transcriptomes from BM cells, blood cells and activated monocytes. As serological markers of translocation, 1,3 beta-glycan, intestinal fatty acid binding protein, and lipopolysaccharide binding protein (LBP) were determined by LAL and ELISA. RESULTS: Transcriptome analysis identified axSpA-specific monocyte signatures showing an imprint of LPS/cytokine-activated monocytes, late granulopoietic BM cells, blood neutrophils, and G-CSF-mobilized blood cells, which suggests LPS/TNF activation and more prominent BM adaptation promoting a neutrophil-like phenotype. GSEA mapped axSpA upregulated genes to inflammatory responses and TNFα signaling and downregulated probe-sets to metabolic pathways. Among translocation markers, LBP levels were significantly increased in axSpA patients vs. HC (p < 0.001). Stratified analysis by disease activity and stage identified an "active disease signature" (BASDAI ≥ 4) with an imprint of LPS/cytokine-activated monocytes and CD16+ monocyte subsets. The "AS signature" (vs. non-radiographic axSpA) showed a reinforced neutrophil-like phenotype due to deprivation of dendritic cell transcripts. CONCLUSIONS: The neutrophil-like phenotype of axSpA monocytes points towards a biased monocytopoiesis from granulocyte-monocyte progenitors. This shift in monocytopoiesis and the LPS/cytokine imprint as well as the elevated LBP levels are indicators of systemic inflammation, which may result from bacterial translocation. The BM adaptation is most prominent in AS patients while disease activity appears to be linked to activation and trafficking of monocytes.


Asunto(s)
Monocitos , Espondiloartritis , Citocinas , Perfilación de la Expresión Génica , Humanos , Espondiloartritis/genética , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA