Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Cell Sci ; 134(18)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34494097

RESUMEN

Lysosomal signaling facilitates the migration of immune cells by releasing Ca2+ to activate the actin-based motor myosin II at the cell rear. However, how the actomyosin cytoskeleton physically associates to lysosomes is unknown. We have previously identified myosin II as a direct interactor of Rab7b, a small GTPase that mediates the transport from late endosomes/lysosomes to the trans-Golgi network (TGN). Here, we show that Rab7b regulates the migration of dendritic cells (DCs) in one- and three-dimensional environments. DCs are immune sentinels that transport antigens from peripheral tissues to lymph nodes to activate T lymphocytes and initiate adaptive immune responses. We found that the lack of Rab7b reduces myosin II light chain phosphorylation and the activation of the transcription factor EB (TFEB), which controls lysosomal signaling and is required for fast DC migration. Furthermore, we demonstrate that Rab7b interacts with the lysosomal Ca2+ channel TRPML1 (also known as MCOLN1), enabling the local activation of myosin II at the cell rear. Taken together, our findings identify Rab7b as the missing physical link between lysosomes and the actomyosin cytoskeleton, allowing control of immune cell migration through lysosomal signaling. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Actomiosina , Lisosomas , Citoesqueleto , Células Dendríticas , Endosomas , Humanos
2.
J Immunol ; 198(7): 2785-2795, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28228559

RESUMEN

Targeting Ags to conventional dendritic cells can enhance Ag-specific immune responses. Although most studies have focused on the induction of T cell responses, the mechanisms by which targeting improves Ab responses are poorly understood. In this study we present data on the use of human XCL1 (hXCL1) and hXCL2 fusion vaccines in a murine model. We show that the human chemokines bound type 1 conventional dendritic cells (cDC1), and that immunization with influenza virus hemagglutinin fused to hXCL1 or hXCL2 induced full protection against influenza challenge. Surprisingly, the hXCL1- and hXCL2-fusion vaccines induced better long-term protection associated with stronger induction of neutralizing Abs, and more Ab-secreting cells in bone marrow. In contrast, murine Xcl1 fusion vaccines induced stronger CD8+ T cell responses compared with hXCL1. Further analysis revealed that although murine Xcl1 fusion vaccines induced chemotaxis and were rapidly endocytosed by cDC1, hXCL1 and hXCL2 fusion vaccines did not induce chemotaxis, were less efficiently endocytosed, and consequently, remained on the surface. This difference may explain the enhanced induction of Abs when targeting Ag to cDC1 using hXCL1 and hXCL2, and suggests that immune responses can be manipulated in directing Abs or T cells based on how efficiently the targeted Ag is endocytosed by the DC.


Asunto(s)
Células Dendríticas/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Vacunas contra la Influenza/inmunología , Receptores Acoplados a Proteínas G/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Quimiotaxis de Leucocito/inmunología , Modelos Animales de Enfermedad , Endocitosis/inmunología , Ensayo de Inmunoadsorción Enzimática , Ensayo de Immunospot Ligado a Enzimas , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/inmunología
3.
Eur J Immunol ; 47(1): 193-205, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27718235

RESUMEN

The cytoplasmic tail of CD45 (ct-CD45) is proteolytically cleaved and released upon activation of human phagocytes. It acts on T cells as an inhibitory, cytokine-like factor in vitro. Here, we show that ct-CD45 is abundant in human peripheral blood plasma from healthy adults compared with plasma derived from umbilical cord blood and plasma from patients with rheumatoid arthritis or systemic lupus erythematosus. Plasma depleted of ct-CD45 enhanced T-cell proliferation, while addition of exogenous ct-CD45 protein inhibited proliferation and reduced cytokine production of human T lymphocytes in response to TCR signaling. Inhibition of T-cell proliferation by ct-CD45 was overcome by costimulation via CD28. T-cell activation in the presence of ct-CD45 was associated with an upregulation of the quiescence factors Schlafen family member 12 (SLFN12) and Krueppel-like factor 2 (KLF2) as well as of the cyclin-dependent kinase (CDK) inhibitor p27kip1. In contrast, positive regulators of the cell cycle such as cyclin D2 and D3 as well as CDK2 and CDK4 were found to be downregulated in response to ct-CD45. In summary, we demonstrate that ct-CD45 is present in human plasma and sets the threshold of T-cell activation.


Asunto(s)
Ciclo Celular , Antígenos Comunes de Leucocito/sangre , Dominios Proteicos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Adulto , Anciano , Artritis Reumatoide/genética , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Biomarcadores , Ciclo Celular/genética , Ciclo Celular/inmunología , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Inmunomodulación , Inmunofenotipificación , Antígenos Comunes de Leucocito/química , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Fenotipo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
4.
Eur J Immunol ; 45(1): 32-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25403892

RESUMEN

Dendritic cells (DCs) are key regulators of both innate and adaptive immunity. During infection, DCs recognise pathogen-associated molecular patterns (PAMPs) via pattern recognition receptors (PRRs) including the Toll-like receptor (TLR) family. TLRs mainly signal via the adaptor protein MyD88. This signalling pathway is required for immune protection during many infections, which are lethal in the absence of MyD88. However, the cell type specific importance of this pathway during both innate and adaptive immune responses against pathogens in vivo remains ill-defined. We discuss recent findings from conditional KO or gain-of-function mouse models targeting TLR/MyD88 signalling pathways in DCs and other myeloid cells during infection. While the general assumption that MyD88-dependent recognition by DCs is essential for inducing protective immunity holds true in some instances, the results surprisingly indicate a much more complex context-dependent requirement for this pathway in DCs and other myeloid or lymphoid cell-types in vivo. Furthermore, we highlight the advantages of Cre-mediated DC targeting approaches and their possible limitations. We also present future perspectives on the development of new genetic mouse models to target distinct DC subsets in vivo. Such models will serve to understand the functional heterogeneity of DCs in vivo.


Asunto(s)
Candidiasis/inmunología , Células Dendríticas/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/inmunología , Toxoplasmosis/inmunología , Inmunidad Adaptativa , Animales , Candida albicans/inmunología , Candidiasis/genética , Candidiasis/microbiología , Células Dendríticas/microbiología , Células Dendríticas/parasitología , Regulación de la Expresión Génica , Inmunidad Innata , Integrasas/genética , Integrasas/metabolismo , Ratones , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/genética , Regiones Promotoras Genéticas , Transducción de Señal/genética , Receptores Toll-Like/genética , Toxoplasma/inmunología , Toxoplasmosis/genética , Toxoplasmosis/parasitología
5.
Blood ; 124(20): 3081-91, 2014 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-25100743

RESUMEN

Multiple subsets of FMS-like tyrosine kinase 3 ligand (FLT3L)-dependent dendritic cells (DCs) control T-cell tolerance and immunity. In mice, Batf3-dependent CD103(+) DCs efficiently enter lymph nodes and cross-present antigens, rendering this conserved DC subset a promising target for tolerance induction or vaccination. However, only limited numbers of CD103(+) DCs can be isolated with current methods. Established bone marrow culture protocols efficiently generate monocyte-derived DCs or produce a mixture of FLT3L-dependent DC subsets. We show that CD103(+) DC development requires prolonged culture time and continuous action of both FLT3L and granulocyte macrophage colony-stimulating factor (GM-CSF), explained by a dual effect of GM-CSF on DC precursors and differentiating CD103(+) DCs. Accordingly, we established a novel method to generate large numbers of CD103(+) DCs (iCD103-DCs) with limited presence of other DC subsets. iCD103-DCs develop in a Batf3- and Irf8-dependent fashion, express a CD8α/CD103 DC gene signature, cross-present cell-associated antigens, and respond to TLR3 stimulation. Thus, iCD103-DCs reflect key features of tissue CD103(+) DCs. Importantly, iCD103-DCs express high levels of CCR7 upon maturation and migrate to lymph nodes more efficiently than classical monocyte-derived DCs. Finally, iCD103-DCs induce T cell-mediated protective immunity in vivo. Our study provides insights into CD103(+) DC development and function.


Asunto(s)
Antígenos CD/inmunología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/inmunología , Células de la Médula Ósea/citología , Técnicas de Cultivo de Célula/métodos , Células Dendríticas/citología , Células Dendríticas/inmunología , Cadenas alfa de Integrinas/inmunología , Proteínas Represoras/inmunología , Animales , Antígenos CD/análisis , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/análisis , Diferenciación Celular , Células Cultivadas , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Inmunidad Celular , Cadenas alfa de Integrinas/análisis , Proteínas de la Membrana/inmunología , Ratones , Proteínas Represoras/análisis , Linfocitos T/inmunología , Receptor Toll-Like 3/inmunología
6.
Eur J Immunol ; 44(5): 1399-409, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24435955

RESUMEN

Tuberculosis is a chronic infectious disease caused by Mycobacterium tuberculosis that is responsible for almost 1.5 million deaths per year. Sensing of mycobacteria by the host's immune system relies on different families of receptors present on innate immune cells. Amongst them, several members of the TLR family are involved in the activation of immune cells by mycobacteria, yet the in vivo contribution of individual TLRs to the protective immune response remains controversial. On the contrary, MyD88, the adaptor molecule for most TLRs, plays a non-redundant role in the protection against tuberculosis and mice with a complete germline deletion of MyD88 succumb very early to infection. MyD88 is expressed in both immune and non-immune cells, but it is not clear whether control of mycobacteria requires ubiquitous or cell-type specific MyD88 expression. Therefore, using novel conditional switch-on mouse models, we aimed to investigate the importance of MyD88 signalling in DCs and macrophages for the induction of protective effector mechanisms against mycobacterial infection. We conclude that specific reactivation of MyD88 signalling in CD11c- or lysozyme M-expressing myeloid cells during Mycobacterium bovis Bacille Calmette-Guerin infection is sufficient to restore systemic and local inflammatory cytokine production and to control pathogen burden.


Asunto(s)
Citocinas/inmunología , Macrófagos/inmunología , Mycobacterium bovis/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Transducción de Señal/inmunología , Tuberculosis/inmunología , Animales , Antígeno CD11c/biosíntesis , Antígeno CD11c/genética , Antígeno CD11c/inmunología , Enfermedad Crónica , Citocinas/biosíntesis , Citocinas/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Modelos Animales de Enfermedad , Eliminación de Gen , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Muramidasa/biosíntesis , Muramidasa/genética , Muramidasa/inmunología , Mycobacterium bovis/metabolismo , Factor 88 de Diferenciación Mieloide/biosíntesis , Factor 88 de Diferenciación Mieloide/genética , Transducción de Señal/genética , Tuberculosis/genética , Tuberculosis/metabolismo , Tuberculosis/patología , Tuberculosis/prevención & control , Tuberculosis/veterinaria
7.
PLoS Pathog ; 9(9): e1003648, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24086137

RESUMEN

Plasmacytoid dendritic cells (pDCs) express the I-type lectin receptor Siglec-H and produce interferon α (IFNα), a critical anti-viral cytokine during the acute phase of murine cytomegalovirus (MCMV) infection. The ligands and biological functions of Siglec-H still remain incompletely defined in vivo. Thus, we generated a novel bacterial artificial chromosome (BAC)-transgenic "pDCre" mouse which expresses Cre recombinase under the control of the Siglec-H promoter. By crossing these mice with a Rosa26 reporter strain, a representative fraction of Siglec-H⁺ pDCs is terminally labeled with red fluorescent protein (RFP). Interestingly, systemic MCMV infection of these mice causes the downregulation of Siglec-H surface expression. This decline occurs in a TLR9- and MyD88-dependent manner. To elucidate the functional role of Siglec-H during MCMV infection, we utilized a novel Siglec-H deficient mouse strain. In the absence of Siglec-H, the low infection rate of pDCs with MCMV remained unchanged, and pDC activation was still intact. Strikingly, Siglec-H deficiency induced a significant increase in serum IFNα levels following systemic MCMV infection. Although Siglec-H modulates anti-viral IFNα production, the control of viral replication was unchanged in vivo. The novel mouse models will be valuable to shed further light on pDC biology in future studies.


Asunto(s)
Células Dendríticas/inmunología , Infecciones por Herpesviridae/inmunología , Interferón-alfa/inmunología , Lectinas/inmunología , Modelos Inmunológicos , Muromegalovirus/fisiología , Células Plasmáticas/inmunología , Receptores de Superficie Celular/inmunología , Animales , Células Dendríticas/metabolismo , Células Dendríticas/patología , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/patología , Interferón-alfa/genética , Interferón-alfa/metabolismo , Lectinas/genética , Lectinas/metabolismo , Ratones , Ratones Noqueados , Células Plasmáticas/metabolismo , Células Plasmáticas/patología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Replicación Viral/genética , Replicación Viral/inmunología
8.
Eur J Immunol ; 43(10): 2543-53, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23784881

RESUMEN

Vaccination is one of the oldest yet still most effective methods to prevent infectious diseases. However, eradication of intracellular pathogens and treatment of certain diseases like cancer requiring efficient cytotoxic immune responses remain a medical challenge. In mice, a successful approach to induce strong cytotoxic CD8⁺ T-cell (CTL) reactions is to target antigens to DCs using specific antibodies against surface receptors in combination with adjuvants. A major drawback for translating this strategy into one for the clinic is the lack of analogous targets in human DCs. DC-SIGN (DC-specific-ICAM3-grabbing-nonintegrin/CD209) is a C-type lectin receptor with potent endocytic capacity and a highly restricted expression on human immature DCs. Therefore, DC-SIGN represents an ideal candidate for DC targeting. Using transgenic mice that express human DC-SIGN under the control of the murine CD11c promoter (hSIGN mice), we explored the efficacy of anti-DC-SIGN antibodies to target antigens to DCs and induce protective immune responses in vivo. We show that anti-DC-SIGN antibodies conjugated to OVA induced strong and persistent antigen-specific CD4⁺ and CD8⁺ T-cell responses, which efficiently protected from infection with OVA-expressing Listeria monocytogenes. Thus, we propose DC targeting via DC-SIGN as a promising strategy for novel vaccination protocols against intracellular pathogens.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Moléculas de Adhesión Celular/metabolismo , Células Dendríticas/inmunología , Lectinas Tipo C/metabolismo , Listeria monocytogenes/inmunología , Receptores de Superficie Celular/metabolismo , Adyuvantes Inmunológicos/genética , Adyuvantes Inmunológicos/metabolismo , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Antígeno CD11c/genética , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Humanos , Inmunidad Activa , Inmunidad Celular , Inmunomodulación , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Listeria monocytogenes/genética , Ratones , Ratones Transgénicos , Ovalbúmina/genética , Ovalbúmina/metabolismo , Regiones Promotoras Genéticas/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Transgenes/genética , Vacunación
9.
Methods Mol Biol ; 2618: 133-145, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36905514

RESUMEN

Dendritic cells (DCs) represent one of the most important immune cell subsets in preventing the host from pathogen invasion by promoting both innate and adaptive immunity. Most research on human dendritic cells has focused on the easy-to-obtain dendritic cells derived in vitro from monocytes (MoDCs). However, many questions remain unanswered regarding the role of different dendritic cell types. The investigation of their roles in human immunity is hampered by their rarity and fragility, which especially holds true for type 1 conventional dendritic cells (cDC1s) and for plasmacytoid dendritic cells (pDCs). In vitro differentiation from hematopoietic progenitors emerged as a common way to produce different DC types, but the efficiency and reproducibility of these protocols needed to be improved and the extent to which the DCs generated in vitro resembled their in vivo counterparts required a more rigorous and global assessment. Here, we describe a cost-effective and robust in vitro differentiation system for the production of cDC1s and pDCs equivalent to their blood counterparts, from cord blood CD34+ hematopoietic stem cells (HSCs) cultured on a stromal feeder layer with a combination of cytokines and growth factors.


Asunto(s)
Células Dendríticas , Células Madre Hematopoyéticas , Humanos , Reproducibilidad de los Resultados , Diferenciación Celular , Antígenos CD34/metabolismo
10.
Eur J Cancer ; 143: 101-112, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33296830

RESUMEN

BACKGROUND: Pancreatic cancer (PC) ranks among the deadliest malignancies worldwide. In the MPACT study, first-line nab-paclitaxel plus gemcitabine (nab-P/G) demonstrated activity (median overall survival [OS], 8.7 months) and tolerability in patients with metastatic PC (mPC). However, the clinical evidence of nab-P/G in the elderly (>70 years), who account for the majority of patients with mPC, is limited. This is the first prospective, multicentre, non-interventional study evaluating the tolerability and effectiveness of nab-P/G in younger (≤70 years) versus elderly (>70 years) patients with mPC in the daily clinical routine. METHODS: Eligible patients with mPC were treated with nab-P/G and observed until disease progression or unacceptable toxicity. The primary objectives were safety and tolerability of nab-P/G, and the secondary objectives were efficacy and real-life dosing. RESULTS: A total of 317 patients with mPC (median age, 70 years) were recruited, of which 299, aged ≤70 (n = 162) and >70 (n = 137) years, were eligible for analysis. Baseline characteristics and the safety profile were comparable between the groups. However, fatigue (22.8% versus 13.0%) and decreased appetite (8.8% versus 1.2%) were more frequent in elderly patients. Younger versus elderly patients equally benefited in terms of objective response rate (36% versus 48%), median progression-free survival (5.6 versus 5.5 months; hazard ratio [HR] = 1.03; p = 0.81) and OS (10.6 versus 10.2 months; HR = 0.89; p = 0.4). In addition, the median treatment duration (5 versus 4 cycles), relative dose intensity (70% versus 74%) or reasons for treatment discontinuation were similar. Most patients (56.2% versus 47.4%) benefited from a second-line therapy. CONCLUSION: This prospective real-world analysis confirms the feasibility and tolerability of nab-P/G treatment and reveals OS data similar for younger patients and elderly patients aged >70 years. CLINICALTRIALS. GOV REGISTRATION: NCT02555813. AUSTRIAN NIS REGISTRY: NIS005071.


Asunto(s)
Albúminas/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Desoxicitidina/análogos & derivados , Paclitaxel/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Anciano , Albúminas/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Femenino , Humanos , Masculino , Paclitaxel/farmacología , Neoplasias Pancreáticas/patología , Gemcitabina
11.
Cell Rep ; 24(7): 1902-1915.e6, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30110645

RESUMEN

The ability to generate large numbers of distinct types of human dendritic cells (DCs) in vitro is critical for accelerating our understanding of DC biology and harnessing them clinically. We developed a DC differentiation method from human CD34+ precursors leading to high yields of plasmacytoid DCs (pDCs) and both types of conventional DCs (cDC1s and cDC2s). The identity of the cells generated in vitro and their strong homology to their blood counterparts were demonstrated by phenotypic, functional, and single-cell RNA-sequencing analyses. This culture system revealed a critical role of Notch signaling and GM-CSF for promoting cDC1 generation. Moreover, we discovered a pre-terminal differentiation state for each DC type, characterized by high expression of cell-cycle genes and lack of XCR1 in the case of cDC1. Our culture system will greatly facilitate the simultaneous and comprehensive study of primary, otherwise rare human DC types, including their mutual interactions.


Asunto(s)
Linaje de la Célula/inmunología , Células Dendríticas/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Receptor Notch1/genética , Antígenos CD34/genética , Antígenos CD34/inmunología , Proteínas de Unión al Calcio , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/inmunología , Diferenciación Celular/efectos de los fármacos , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Expresión Génica , Perfilación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Imidazoles/farmacología , Inmunofenotipificación , Péptidos y Proteínas de Señalización Intercelular/inmunología , Lipopolisacáridos/farmacología , Proteínas de la Membrana/inmunología , Poli I-C/farmacología , Cultivo Primario de Células , Receptor Notch1/inmunología , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/inmunología , Transducción de Señal , Análisis de la Célula Individual
12.
Front Immunol ; 9: 495, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29675017

RESUMEN

Mycobacterium tuberculosis (Mtb), the causative agent of human tuberculosis, is able to efficiently manipulate the host immune system establishing chronic infection, yet the underlying mechanisms of immune evasion are not fully understood. Evidence suggests that this pathogen interferes with host cell lipid metabolism to ensure its persistence. Fatty acid metabolism is regulated by acetyl-CoA carboxylase (ACC) 1 and 2; both isoforms catalyze the conversion of acetyl-CoA into malonyl-CoA, but have distinct roles. ACC1 is located in the cytosol, where it regulates de novo fatty acid synthesis (FAS), while ACC2 is associated with the outer mitochondrial membrane, regulating fatty acid oxidation (FAO). In macrophages, mycobacteria induce metabolic changes that lead to the cytosolic accumulation of lipids. This reprogramming impairs macrophage activation and contributes to chronic infection. In dendritic cells (DCs), FAS has been suggested to underlie optimal cytokine production and antigen presentation, but little is known about the metabolic changes occurring in DCs upon mycobacterial infection and how they affect the outcome of the immune response. We therefore determined the role of fatty acid metabolism in myeloid cells and T cells during Mycobacterium bovis BCG or Mtb infection, using novel genetic mouse models that allow cell-specific deletion of ACC1 and ACC2 in DCs, macrophages, or T cells. Our results demonstrate that de novo FAS is induced in DCs and macrophages upon M. bovis BCG infection. However, ACC1 expression in DCs and macrophages is not required to control mycobacteria. Similarly, absence of ACC2 did not influence the ability of DCs and macrophages to cope with infection. Furthermore, deletion of ACC1 in DCs or macrophages had no effect on systemic pro-inflammatory cytokine production or T cell priming, suggesting that FAS is dispensable for an intact innate response against mycobacteria. In contrast, mice with a deletion of ACC1 specifically in T cells fail to generate efficient T helper 1 responses and succumb early to Mtb infection. In summary, our results reveal ACC1-dependent FAS as a crucial mechanism in T cells, but not DCs or macrophages, to fight against mycobacterial infection.


Asunto(s)
Células Dendríticas/inmunología , Ácidos Grasos/inmunología , Inmunidad Innata , Macrófagos/inmunología , Mycobacterium tuberculosis/inmunología , Células TH1/inmunología , Tuberculosis/inmunología , Acetil-CoA Carboxilasa/genética , Acetil-CoA Carboxilasa/inmunología , Animales , Células Dendríticas/microbiología , Células Dendríticas/patología , Ácidos Grasos/genética , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Noqueados , Mycobacterium bovis/inmunología , Mycobacterium tuberculosis/genética , Células TH1/microbiología , Células TH1/patología , Tuberculosis/genética , Tuberculosis/patología
13.
Cell Metab ; 28(3): 504-515.e7, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30043753

RESUMEN

T cell subsets including effector (Teff), regulatory (Treg), and memory (Tmem) cells are characterized by distinct metabolic profiles that influence their differentiation and function. Previous research suggests that engagement of long-chain fatty acid oxidation (LC-FAO) supports Foxp3+ Treg cell and Tmem cell survival. However, evidence for this is mostly based on inhibition of Cpt1a, the rate-limiting enzyme for LC-FAO, with the drug etomoxir. Using genetic models to target Cpt1a specifically in T cells, we dissected the role of LC-FAO in primary, memory, and regulatory T cell responses. Here we show that the ACC2/Cpt1a axis is largely dispensable for Teff, Tmem, or Treg cell formation, and that the effects of etomoxir on T cell differentiation and function are independent of Cpt1a expression. Together our data argue that metabolic pathways other than LC-FAO fuel Tmem or Treg differentiation and suggest alternative mechanisms for the effects of etomoxir that involve mitochondrial respiration.


Asunto(s)
Acetil-CoA Carboxilasa/fisiología , Linfocitos T CD8-positivos/metabolismo , Carnitina O-Palmitoiltransferasa/fisiología , Compuestos Epoxi/farmacología , Ácidos Grasos/metabolismo , Memoria Inmunológica/efectos de los fármacos , Mitocondrias/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Acetil-CoA Carboxilasa/genética , Animales , Carnitina O-Palmitoiltransferasa/genética , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Niño , Preescolar , Femenino , Técnicas de Inactivación de Genes , Humanos , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , Linfocitos T Reguladores/metabolismo
14.
Cell Rep ; 6(4): 698-708, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24529704

RESUMEN

Listeria monocytogenes (LM), a facultative intracellular Gram-positive pathogen, can cause life-threatening infections in humans. In mice, the signaling cascade downstream of the myeloid differentiation factor 88 (MyD88) is essential for proper innate immune activation against LM, as MyD88-deficient mice succumb early to infection. Here, we show that MyD88 signaling in dendritic cells (DCs) is sufficient to mediate the protective innate response, including the production of proinflammatory cytokines, neutrophil infiltration, bacterial clearance, and full protection from lethal infection. We also demonstrate that MyD88 signaling by DCs controls the infection rates of CD8α(+) cDCs and thus limits the spread of LM to the T cell areas. Furthermore, in mice expressing MyD88 in DCs, inflammatory monocytes, which are required for bacterial clearance, are activated independently of intrinsic MyD88 signaling. In conclusion, CD11c(+) conventional DCs critically integrate pathogen-derived signals via MyD88 signaling during early infection with LM in vivo.


Asunto(s)
Células Dendríticas/metabolismo , Inmunidad Innata , Listeriosis/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Animales , Antígeno CD11c/genética , Antígeno CD11c/metabolismo , Antígenos CD8/genética , Antígenos CD8/metabolismo , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/inmunología , Listeriosis/metabolismo , Ratones , Factor 88 de Diferenciación Mieloide/genética , Neutrófilos/inmunología , Transducción de Señal , Linfocitos T/inmunología
15.
Science ; 338(6106): 532-6, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-23112334

RESUMEN

Although regulatory T cells (T(regs)) are known to suppress self-reactive autoimmune responses, their role during T cell responses to nonself antigens is not well understood. We show that T(regs) play a critical role during the priming of immune responses in mice. T(reg) depletion induced the activation and expansion of a population of low-avidity CD8(+) T cells because of overproduction of CCL-3/4/5 chemokines, which stabilized the interactions between antigen-presenting dendritic cells and low-avidity T cells. In the absence of T(regs), the avidity of the primary immune response was impaired, which resulted in reduced memory to Listeria monocytogenes. These results suggest that T(regs) are important regulators of the homeostasis of CD8(+) T cell priming and play a critical role in the induction of high-avidity primary responses and effective memory.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunidad Celular , Memoria Inmunológica , Linfocitos T Reguladores/inmunología , Animales , Quimiocina CCL1/metabolismo , Quimiocina CCL4/metabolismo , Quimiocina CCL5/metabolismo , Femenino , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor , Proteínas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA