Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros











Intervalo de año de publicación
1.
Curr Opin Immunol ; 89: 102454, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39154521

RESUMEN

TCF-1+ CD8+ T cell populations have emerged as critical determinants for long-lived immunological memory. This cell population has stem-like properties and is implicated in improved disease outcomes by driving sustained killing of infected cells and maintaining the immune-cancer equilibrium. During an immune response, several factors, including antigen deposition and affinity, the inflammatory milieu, and T cell priming dynamics, aggregate to skew CD8+ T cell differentiation. Although these mechanisms are altered between acute and chronic disease settings, phenotypically similar stem-like TCF-1+ CD8+ T cell states are formed in each of these settings. Here, we characterize the specialized microenvironments within lymph nodes and the tumor microenvironment, which foster the generation or re-activation of stem-like TCF-1+ CD8+ T cell populations. We highlight the potential for targeting the stem-like CD8+ T cell niche to enhance vaccination and cancer immunotherapy and to track the trajectory of stem-like CD8+ T cells as biomarkers of therapeutic efficacy.


Asunto(s)
Linfocitos T CD8-positivos , Microambiente Tumoral , Linfocitos T CD8-positivos/inmunología , Humanos , Animales , Microambiente Tumoral/inmunología , Diferenciación Celular/inmunología , Memoria Inmunológica , Neoplasias/inmunología , Neoplasias/terapia , Nicho de Células Madre/inmunología , Ganglios Linfáticos/inmunología
2.
bioRxiv ; 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38979239

RESUMEN

Developing vaccines that promote CD8 + T cell memory is a challenge for infectious disease and cancer immunotherapy. TCF-1 + stem cell-like memory T (T SCM ) cells are important determinants of long-lived memory. Yet, the developmental requirements for T SCM formation are unclear. Here, we identify the temporal window for type I interferon (IFN-I) receptor (IFNAR) blockade to drive T SCM cell generation. T SCM cells were transcriptionally distinct and emerged from a transitional precursor of exhausted (T PEX ) cellular state concomitant with viral clearance. T SCM differentiation correlated with T cell retention within the lymph node paracortex, due to increased CXCR3 chemokine abundance which disrupted gradient formation. These affects were due a counterintuitive increase in IFNψ, which controlled cell location. Combining IFNAR inhibition with mRNA-LNP vaccination promoted specific T SCM differentiation and enhanced protection against chronic infection. These finding propose a new approach to vaccine design whereby modulation of inflammation promotes memory formation and function. HIGHLIGHTS: Early, transient inhibition of the type I interferon (IFN) receptor (IFNAR) during acute viral infection promotes stem cell-like memory T (T SCM ) cell differentiation without establishing chronic infection. T SCM and precursor of exhausted (T PEX ) cellular states are distinguished transcriptionally and by cell surface markers. Developmentally, T SCM cell differentiation occurs via a transition from a T PEX state coinciding with viral clearance. Transient IFNAR blockade increases IFNψ production to modulate the ligands of CXCR3 and couple T SCM differentiation to cell retention within the T cell paracortex of the lymph node. Specific promotion of T SCM cell differentiation with nucleoside-modified mRNA-LNP vaccination elicits enhanced protection against chronic viral challenge.

3.
Immunity ; 57(5): 1037-1055.e6, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38593796

RESUMEN

Memory B cells (MBCs) are key providers of long-lived immunity against infectious disease, yet in chronic viral infection, they do not produce effective protection. How chronic viral infection disrupts MBC development and whether such changes are reversible remain unknown. Through single-cell (sc)ATAC-seq and scRNA-seq during acute versus chronic lymphocytic choriomeningitis viral infection, we identified a memory subset enriched for interferon (IFN)-stimulated genes (ISGs) during chronic infection that was distinct from the T-bet+ subset normally associated with chronic infection. Blockade of IFNAR-1 early in infection transformed the chromatin landscape of chronic MBCs, decreasing accessibility at ISG-inducing transcription factor binding motifs and inducing phenotypic changes in the dominating MBC subset, with a decrease in the ISG subset and an increase in CD11c+CD80+ cells. However, timing was critical, with MBCs resistant to intervention at 4 weeks post-infection. Together, our research identifies a key mechanism to instruct MBC identity during viral infection.


Asunto(s)
Epigénesis Genética , Interferón Tipo I , Coriomeningitis Linfocítica , Virus de la Coriomeningitis Linfocítica , Células B de Memoria , Animales , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Ratones , Virus de la Coriomeningitis Linfocítica/inmunología , Células B de Memoria/inmunología , Ratones Endogámicos C57BL , Receptor de Interferón alfa y beta/genética , Memoria Inmunológica/inmunología , Enfermedad Crónica , Subgrupos de Linfocitos B/inmunología , Análisis de la Célula Individual
4.
Nat Immunol ; 25(2): 330-342, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38172260

RESUMEN

Antibody-secreting plasma cells (PCs) are generated in secondary lymphoid organs but are reported to reside in an emerging range of anatomical sites. Analysis of the transcriptome of different tissue-resident (Tr)PC populations revealed that they each have their own transcriptional signature indicative of functional adaptation to the host tissue environment. In contrast to expectation, all TrPCs were extremely long-lived, regardless of their organ of residence, with longevity influenced by intrinsic factors like the immunoglobulin isotype. Analysis at single-cell resolution revealed that the bone marrow is unique in housing a compendium of PCs generated all over the body that retain aspects of the transcriptional program indicative of their tissue of origin. This study reveals that extreme longevity is an intrinsic property of TrPCs whose transcriptome is imprinted by signals received both at the site of induction and within the tissue of residence.


Asunto(s)
Médula Ósea , Células Plasmáticas , Células de la Médula Ósea
5.
Immunol Cell Biol ; 101(4): 345-357, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36710659

RESUMEN

The transcription factor Myc is critically important in driving cell proliferation, a function that is frequently dysregulated in cancer. To avoid this dysregulation Myc is tightly controlled by numerous layers of regulation. One such layer is the use of distal regulatory enhancers to drive Myc expression. Here, using chromosome conformation capture to examine B cells of the immune system in the first hours after their activation, we reveal a previously unidentified enhancer of Myc. The interactivity of this enhancer coincides with a dramatic, but discrete, spike in Myc expression 3 h post-activation. However, genetic deletion of this region, has little impact on Myc expression, Myc protein level or in vitro and in vivo cell proliferation. Examination of the enhancer deleted regulatory landscape suggests that enhancer redundancy likely sustains Myc expression. This work highlights not only the importance of temporally examining enhancers, but also the complexity and dynamics of the regulation of critical genes such as Myc.


Asunto(s)
Elementos de Facilitación Genéticos , Genes myc , Elementos de Facilitación Genéticos/genética , Factores de Transcripción/metabolismo , Regulación de la Expresión Génica , Regiones Promotoras Genéticas
6.
Cell Death Differ ; 29(12): 2519-2530, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35831623

RESUMEN

High-throughput methodologies are the cornerstone of screening approaches to identify novel compounds that regulate immune cell function. To identify novel targeted therapeutics to treat immune disorders and haematological malignancies, there is a need to integrate functional cellular information with the molecular mechanisms that regulate changes in immune cell phenotype. We facilitate this goal by combining quantitative methods for dissecting complex simultaneous cell phenotypic effects with genomic analysis. This combination strategy we term Multiplexed Analysis of Cells sequencing (MAC-seq), a modified version of Digital RNA with perturbation of Genes (DRUGseq). We applied MAC-seq to screen compounds that target the epigenetic machinery of B cells and assess altered humoral immunity by measuring changes in proliferation, survival, differentiation and transcription. This approach revealed that polycomb repressive complex 2 (PRC2) inhibitors promote antibody secreting cell (ASC) differentiation in both murine and human B cells in vitro. This is further validated using T cell-dependent immunization in mice. Functional dissection of downstream effectors of PRC2 using arrayed CRISPR screening uncovered novel regulators of B cell differentiation, including Mybl1, Myof, Gas7 and Atoh8. Together, our findings demonstrate that integrated phenotype-transcriptome analyses can be effectively combined with drug screening approaches to uncover the molecular circuitry that drives lymphocyte fate decisions.


Asunto(s)
Linfocitos B , Epigénesis Genética , Animales , Humanos , Ratones , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Perfilación de la Expresión Génica , Fenotipo , Complejo Represivo Polycomb 2/metabolismo
7.
Immunity ; 55(3): 423-441.e9, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35139355

RESUMEN

Cell death plays an important role during pathogen infections. Here, we report that interferon-γ (IFNγ) sensitizes macrophages to Toll-like receptor (TLR)-induced death that requires macrophage-intrinsic death ligands and caspase-8 enzymatic activity, which trigger the mitochondrial apoptotic effectors, BAX and BAK. The pro-apoptotic caspase-8 substrate BID was dispensable for BAX and BAK activation. Instead, caspase-8 reduced pro-survival BCL-2 transcription and increased inducible nitric oxide synthase (iNOS), thus facilitating BAX and BAK signaling. IFNγ-primed, TLR-induced macrophage killing required iNOS, which licensed apoptotic caspase-8 activity and reduced the BAX and BAK inhibitors, A1 and MCL-1. The deletion of iNOS or caspase-8 limited SARS-CoV-2-induced disease in mice, while caspase-8 caused lethality independent of iNOS in a model of hemophagocytic lymphohistiocytosis. These findings reveal that iNOS selectively licenses programmed cell death, which may explain how nitric oxide impacts disease severity in SARS-CoV-2 infection and other iNOS-associated inflammatory conditions.


Asunto(s)
COVID-19/inmunología , Caspasa 8/metabolismo , Interferón gamma/metabolismo , Linfohistiocitosis Hemofagocítica/inmunología , Macrófagos/inmunología , Mitocondrias/metabolismo , SARS-CoV-2/fisiología , Animales , Caspasa 8/genética , Células Cultivadas , Citotoxicidad Inmunológica , Humanos , Interferón gamma/genética , Activación de Macrófagos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Transducción de Señal , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
8.
Immunol Rev ; 306(1): 76-92, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34882817

RESUMEN

The lymph node plays a critical role in mounting an adaptive immune response to infection, clearance of foreign pathogens, and cancer immunosurveillance. Within this complex structure, intranodal migration is vital for CD8+ T cell activation and differentiation. Combining tissue clearing and volumetric light sheet fluorescent microscopy of intact lymph nodes has allowed us to explore the spatial regulation of T cell fates. This has determined that short-lived effector (TSLEC ) are imprinted in peripheral lymph node interfollicular regions, due to CXCR3 migration. In contrast, stem-like memory cell (TSCM ) differentiation is determined in the T cell paracortex. Here, we detail the inflammatory and chemokine regulators of spatially restricted T cell differentiation, with a focus on how to promote TSCM . We propose a default pathway for TSCM differentiation due to CCR7-directed segregation of precursors away from the inflammatory effector niche. Although volumetric imaging has revealed the consequences of intranodal migration, we still lack knowledge of how this is orchestrated within a complex chemokine environment. Toward this goal, we highlight the potential of combining microfluidic chambers with pre-determined complexity and subcellular resolution microscopy.


Asunto(s)
Linfocitos T CD8-positivos , Memoria Inmunológica , Diferenciación Celular , Quimiocinas/metabolismo , Humanos , Ganglios Linfáticos , Activación de Linfocitos
9.
Nat Immunol ; 22(7): 851-864, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099918

RESUMEN

Group 2 innate lymphoid cells (ILC2s) are essential to maintain tissue homeostasis. In cancer, ILC2s can harbor both pro-tumorigenic and anti-tumorigenic functions, but we know little about their underlying mechanisms or whether they could be clinically relevant or targeted to improve patient outcomes. Here, we found that high ILC2 infiltration in human melanoma was associated with a good clinical prognosis. ILC2s are critical producers of the cytokine granulocyte-macrophage colony-stimulating factor, which coordinates the recruitment and activation of eosinophils to enhance antitumor responses. Tumor-infiltrating ILC2s expressed programmed cell death protein-1, which limited their intratumoral accumulation, proliferation and antitumor effector functions. This inhibition could be overcome in vivo by combining interleukin-33-driven ILC2 activation with programmed cell death protein-1 blockade to significantly increase antitumor responses. Together, our results identified ILC2s as a critical immune cell type involved in melanoma immunity and revealed a potential synergistic approach to harness ILC2 function for antitumor immunotherapies.


Asunto(s)
Anticuerpos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Inhibidores de Puntos de Control Inmunológico/farmacología , Interleucina-33/farmacología , Linfocitos/efectos de los fármacos , Melanoma Experimental/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Quimiotaxis de Leucocito/efectos de los fármacos , Citotoxicidad Inmunológica/efectos de los fármacos , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Eosinófilos/metabolismo , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Linfocitos/inmunología , Linfocitos/metabolismo , Masculino , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo
10.
Immunol Rev ; 300(1): 203-219, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33586207

RESUMEN

The relationship between the extrinsic environment and the internal transcriptional network is circular. Naive T cells first engage with antigen-presenting cells to set transcriptional differentiation networks in motion. In turn, this regulates specific chemokine receptors that direct migration into distinct lymph node niches. Movement into these regions brings newly activated T cells into contact with accessory cells and cytokines that reinforce the differentiation programming to specify T cell function. We and others have observed similarities in the transcriptional networks that specify both CD4+ T follicular helper (TFH ) cells and CD8+ central memory stem-like (TSCM ) cells. Here, we compare and contrast the current knowledge for these shared differentiation programs, compared to their effector counterparts, CD4+ T-helper 1 (TH1 ) and CD8+ short-lived effector (TSLEC ) cells. Understanding the interplay between cellular interactions and transcriptional programming is essential to harness T cell differentiation that is fit for purpose; to stimulate potent T cell effector function for the elimination of chronic infection and cancer; or to amplify the formation of humoral immunity and longevity of cellular memory to prevent infectious diseases.


Asunto(s)
Comunicación Celular , Linfocitos T , Células Presentadoras de Antígenos , Diferenciación Celular , Receptores de Quimiocina , Linfocitos T Colaboradores-Inductores
11.
Cell Rep ; 33(11): 108504, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33326791

RESUMEN

Histone modifiers are essential for the ability of immune cells to reprogram their gene expression during differentiation. The recruitment of the histone methyltransferase DOT1L (disruptor of telomeric silencing 1-like) induces oncogenic gene expression in a subset of B cell leukemias. Despite its importance, its role in the humoral immune system is unclear. Here, we demonstrate that DOT1L is a critical regulator of B cell biology. B cell development is defective in Dot1lf/fMb1Cre/+ mice, culminating in a reduction of peripheral mature B cells. Upon immunization or influenza infection of Dot1lf/fCd23Cre/+ mice, class-switched antibody-secreting cells are significantly attenuated and germinal centers fail to form. Consequently, DOT1L is essential for B cell memory formation. Transcriptome, pathway, and histological analyses identified a role for DOT1L in reprogramming gene expression for appropriate localization of B cells during the initial stage of the response. Together, these results demonstrate an essential role for DOT1L in generating an effective humoral immune response.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/inmunología , Inmunidad Humoral/inmunología , Metiltransferasas/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones
12.
Immunol Rev ; 289(1): 101-114, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30977199

RESUMEN

A fundamental question in immunology is how cells decide between distinct T helper, effector or memory differentiation fates. These decisions are paramount to overcome infection and establish long-lasting protection. The impact of cell location for the determination of T-cell fate decisions is an emerging field. This review will discuss our current understanding of the migration path that T cells follow, within draining lymph nodes, to steer differentiation down distinct paths of either effector or memory fates. In particular, the regulation of migration and cellular encounters mediated by the chemokine receptor CXCR3 and its ligands will be discussed. The combination of increased antigen density and unique cellular partners play a central role in facilitating the site-specific differentiation of effector T cells, within the interfollicular regions of draining lymph nodes. Recent advances have applied this knowledge to optimize vaccine design to target antigen to lymph nodes. Increased understanding of the regulation of CXCR3 ligands and how T cells integrate multiple chemokine cues will help further progress in this field and allow additional applications to direct cell differentiation outside the lymph node, to enhance memory residency in peripheral tissues and effector anti-tumor responses.


Asunto(s)
Quimiocinas/metabolismo , Receptores CXCR3/metabolismo , Linfocitos T/inmunología , Animales , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Humanos , Inmunomodulación , Activación de Linfocitos
13.
J Exp Med ; 216(3): 621-637, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30723095

RESUMEN

Ectopic lymphoid structures form in a wide range of inflammatory conditions, including infection, autoimmune disease, and cancer. In the context of infection, this response can be beneficial for the host: influenza A virus infection-induced pulmonary ectopic germinal centers give rise to more broadly cross-reactive antibody responses, thereby generating cross-strain protection. However, despite the ubiquity of ectopic lymphoid structures and their role in both health and disease, little is known about the mechanisms by which inflammation is able to convert a peripheral tissue into one that resembles a secondary lymphoid organ. Here, we show that type I IFN produced after viral infection can induce CXCL13 expression in a phenotypically distinct population of lung fibroblasts, driving CXCR5-dependent recruitment of B cells and initiating ectopic germinal center formation. This identifies type I IFN as a novel inducer of CXCL13, which, in combination with other stimuli, can promote lung remodeling, converting a nonlymphoid tissue into one permissive to functional tertiary lymphoid structure formation.


Asunto(s)
Quimiocina CXCL13/metabolismo , Centro Germinal/patología , Interferón Tipo I/metabolismo , Infecciones por Orthomyxoviridae/patología , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Quimiocina CXCL13/genética , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/virología , Centro Germinal/efectos de los fármacos , Centro Germinal/metabolismo , Interferón beta/farmacología , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Orthomyxoviridae/metabolismo , Receptores CXCR5/genética , Receptores CXCR5/metabolismo , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología
14.
Mol Immunol ; 115: 56-63, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30144957

RESUMEN

NK cells are cytotoxic lymphocytes with a key role in limiting tumour metastases. In mice, the NK cell lineage continually expresses high levels of the Inhibitor of DNA-binding 2 (Id2) protein and loss of Id2 is incongruous with their survival due to aberrant E-protein target gene activity. Using novel Id2 and E-protein antibodies that detect both mouse and human proteins, we have extensively characterised Id2 and E-protein expression in murine and human NK cells. We detected clear expression of E2 A and HEB, and to a lesser extent E2-2 in murine NK cells. In contrast HEB appears to be the major E-protein expressed in human NK cells, with minor E2-2 expression and surprisingly, no E2 A detected in primary NK cells nor human NK cell lines. These novel antibodies are also functional in immunofluorescence and immunoprecipitation. Mass spectrometry analysis of Id2 immuno-precipitated from murine NK cells revealed a number of novel associated proteins including several members of the SWI/SNF-related matrix-associated actin-dependent regulator chromatin (SMARC) and Mediator complex (MED) families. Taken together, these data highlight the utility of novel Id2 and E-protein antibodies and caution against mouse models for understanding Id2/E-protein biology in NK cells given their clearly disparate expression patternbetween species.


Asunto(s)
Anticuerpos/inmunología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Proteína 2 Inhibidora de la Diferenciación/inmunología , Células Asesinas Naturales/inmunología , Factor de Transcripción 4/inmunología , Animales , Línea Celular , Linaje de la Célula/inmunología , Cromatina/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL
15.
Front Immunol ; 9: 2454, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30405637

RESUMEN

The skin represents a specialized site for immune surveillance consisting of resident, inflammatory and memory populations of lymphocytes. The entry and retention of T cells, B cells, and ILCs is tightly regulated to facilitate detection of pathogens, inflammation and tumors cells. Loss of individual or multiple populations in the skin may break tolerance or increase susceptibility to tumor growth and spread. Studies have significantly advanced our understanding of the role of skin T cells and ILCs at steady state and in inflammatory settings such as viral challenge, atopy, and autoimmune inflammation. The knowledge raised by these studies can benefit to our understanding of immune cell trafficking in primary melanoma, shedding light on the mechanisms of tumor immune surveillance and to improve immunotherapy. This review will focus on the T cells, B cells, and ILCs of the skin at steady state, in inflammatory context and in melanoma. In particular, we will detail the core chemokine and adhesion molecules that regulate cell trafficking to and within the skin, which may provide therapeutic avenues to promote tumor homing for a team of lymphocytes.


Asunto(s)
Linfocitos B/inmunología , Movimiento Celular/inmunología , Melanoma/patología , Piel/citología , Piel/inmunología , Linfocitos T/inmunología , Quimiocinas/metabolismo , Humanos , Inflamación/inmunología , Integrinas/metabolismo , Melanoma/inmunología , Selectinas/metabolismo , Piel/patología
16.
Front Immunol ; 9: 482, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29568300

RESUMEN

T-dependent humoral immune responses to infection involve a collaboration between B and CD4 T cell activation, migration, and co-stimulation, thereby culminating in the formation of germinal centers (GCs) and eventual differentiation into memory cells and long-lived plasma cells (PCs). CD4 T cell-derived signals drive the formation of a tailored B cell response. Downstream of these signals are transcriptional regulators that are the critical enactors of immune cell programs. In particular, a core group of transcription factors regulate both B and T cell differentiation, identity, and function. The timing and expression levels of these transcription factors are tightly controlled, with dysregulated expression correlated to immune cell dysfunction in autoimmunity and lymphomagenesis. Recent studies have significantly advanced our understanding of both extrinsic and intrinsic regulators of autoreactive B cells and antibody-secreting PCs in systemic lupus erythematosus, rheumatoid arthritis, and other autoimmune conditions. Yet, there are still gaps in our understanding of the causative role these regulators play, as well as the link between lymphoid responses and peripheral damage. This review will focus on the genesis of immunopathogenic CD4 helper and GC B cells. In particular, we will detail the transcriptional regulation of cytokine and chemokine receptor signaling during the pathogenesis of GC-derived autoimmune conditions in both murine models and human patients.


Asunto(s)
Autoinmunidad , Diferenciación Celular/inmunología , Lupus Eritematoso Sistémico/inmunología , Células Plasmáticas/inmunología , Transducción de Señal/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Factores de Transcripción/inmunología , Animales , Autoanticuerpos/inmunología , Humanos , Lupus Eritematoso Sistémico/patología , Células Plasmáticas/patología , Linfocitos T Colaboradores-Inductores/patología
17.
Cell Rep ; 19(3): 461-470, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28423310

RESUMEN

Humoral immune responses are tailored to the invading pathogen through regulation of key transcription factors and their networks. This is critical to establishing effective antibody-mediated responses, yet it is unknown how B cells integrate pathogen-induced signals to drive or suppress transcriptional programs specialized for each class of pathogen. Here, we detail the key role of the transcription factor c-Myb in regulating the T-bet-mediated anti-viral program. Deletion of c-Myb in mature B cells significantly increased serum IgG2c and CXCR3 expression by upregulating T-bet, normally suppressed during Th2-cell-mediated responses. Enhanced expression of T-bet resulted in aberrant plasma cell differentiation within the germinal center, mediated by CXCR3 expression. These findings identify a dual role for c-Myb in limiting inappropriate effector responses while coordinating plasma cell differentiation with germinal center egress. Identifying such intrinsic regulators of specialized antibody responses can assist in vaccine design and therapeutic intervention in B-cell-mediated immune disorders.


Asunto(s)
Formación de Anticuerpos/inmunología , Linfocitos B/citología , Linfocitos B/inmunología , Diferenciación Celular , Proteínas Proto-Oncogénicas c-myb/metabolismo , Proteínas de Dominio T Box/metabolismo , Animales , Afinidad de Anticuerpos , Femenino , Eliminación de Gen , Regulación de la Expresión Génica , Centro Germinal/citología , Centro Germinal/metabolismo , Humanos , Masculino , Ratones , Células Plasmáticas/citología , Células Plasmáticas/metabolismo , Proteínas Proto-Oncogénicas c-myb/deficiencia , Receptores CXCR3/metabolismo , Sindecano-1/metabolismo , Transcripción Genética
19.
Blood ; 123(22): 3420-8, 2014 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-24723679

RESUMEN

Interleukin-10 (IL-10) is a key immunoregulatory cytokine that functions to prevent inflammatory and autoimmune diseases. Despite the critical role for IL-10 produced by effector CD8(+) T cells during pathogen infection and autoimmunity, the mechanisms regulating its production are poorly understood. We show that loss of the inhibitor of DNA binding 2 (Id2) in T cells resulted in aberrant IL-10 expression in vitro and in vivo during influenza virus infection and in a model of acute graft-versus-host disease (GVHD). Furthermore, IL-10 overproduction substantially reduced the immunopathology associated with GVHD. We demonstrate that Id2 acts to repress the E2A-mediated trans-activation of the Il10 locus. Collectively, our findings uncover a key regulatory role of Id2 during effector T cell differentiation necessary to limit IL-10 production by activated T cells and minimize their suppressive activity during the effector phase of disease control.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Interleucina-10/genética , Subgrupos de Linfocitos T/metabolismo , Activación Transcripcional , Animales , Médula Ósea/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Epigénesis Genética , Regulación de la Expresión Génica , Sitios Genéticos , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/metabolismo , Enfermedad Injerto contra Huésped/mortalidad , Proteína 2 Inhibidora de la Diferenciación/deficiencia , Proteína 2 Inhibidora de la Diferenciación/genética , Interleucina-10/metabolismo , Ratones , Ratones Noqueados , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/mortalidad
20.
Immunity ; 37(6): 1091-103, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23123063

RESUMEN

Differentiation of naive CD4(+) T cells into T helper (Th) cells is a defining event in adaptive immunity. The cytokines and transcription factors that control Th cell differentiation are understood, but it is not known how this process is orchestrated within lymph nodes (LNs). Here we have shown that the CXCR3 chemokine receptor was required for optimal generation of interferon-γ (IFN-γ)-secreting Th1 cells in vivo. By using a CXCR3 ligand reporter mouse, we found that stromal cells predominately expressed the chemokine ligand CXCL9 whereas hematopoietic cells expressed CXCL10 in LNs. Dendritic cell (DC)-derived CXCL10 facilitated T cell-DC interactions in LNs during T cell priming while both chemokines guided intranodal positioning of CD4(+) T cells to interfollicular and medullary zones. Thus, different chemokines acting on the same receptor can function locally to facilitate DC-T cell interactions and globally to influence intranodal positioning, and both functions contribute to Th1 cell differentiation.


Asunto(s)
Diferenciación Celular/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Receptores CXCR3/metabolismo , Células TH1/citología , Células TH1/inmunología , Animales , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Quimiocina CXCL9/genética , Quimiocina CXCL9/inmunología , Quimiocinas CXC/genética , Quimiocinas CXC/inmunología , Citocinas/biosíntesis , Proteínas de Unión al ADN/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Regulación de la Expresión Génica , Interferón gamma/biosíntesis , Ligandos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/metabolismo , Ratones , Ratones Transgénicos , Unión Proteica , Receptores CXCR3/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA