Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Immunohorizons ; 7(11): 737-746, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37934060

RESUMEN

TH17 cells are implicated in the pathogenesis of multiple sclerosis and experimental autoimmune encephalomyelitis (EAE). We previously reported that the transcription factor basic helix-loop-helix family member e40 (BHLHE40) marks cytokine-producing pathogenic TH cells during EAE, and that its expression in T cells is required for clinical disease. In this study, using dual reporter mice, we show BHLHE40 expression within TH1/17 and ex-TH17 cells following EAE induction. Il17a-Cre-mediated deletion of BHLHE40 in TH cells led to less severe EAE with reduced TH cell cytokine production. Characterization of the leukocytes in the CNS during EAE by single-cell RNA sequencing identified differences in the infiltrating myeloid cells when BHLHE40 was present or absent in TH17 cells. Our studies highlight the importance of BHLHE40 in promoting TH17 cell encephalitogenicity and instructing myeloid cell responses during active EAE.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Encefalomielitis Autoinmune Experimental , Células Th17 , Animales , Ratones , Reacciones Cruzadas , Citocinas , Células Mieloides , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo
2.
J Clin Invest ; 133(19)2023 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-37781916

RESUMEN

The comprehensive assessment of long-term effects of reducing intake of energy (CALERIE-II; NCT00427193) clinical trial established that caloric restriction (CR) in humans lowers inflammation. The identity and mechanism of endogenous CR-mimetics that can be deployed to control obesity-associated inflammation and diseases are not well understood. Our studies have found that 2 years of 14% sustained CR in humans inhibits the expression of the matricellular protein, secreted protein acidic and rich in cysteine (SPARC), in adipose tissue. In mice, adipose tissue remodeling caused by weight loss through CR and low-protein diet feeding decreased, while high-fat diet-induced (HFD-induced) obesity increased SPARC expression in adipose tissue. Inducible SPARC downregulation in adult mice mimicked CR's effects on lowering adiposity by regulating energy expenditure. Deletion of SPARC in adipocytes was sufficient to protect mice against HFD-induced adiposity, chronic inflammation, and metabolic dysfunction. Mechanistically, SPARC activates the NLRP3 inflammasome at the priming step and downregulation of SPARC lowers macrophage inflammation in adipose tissue, while excess SPARC activated macrophages via JNK signaling. Collectively, reduction of adipocyte-derived SPARC confers CR-like metabolic and antiinflammatory benefits in obesity by serving as an immunometabolic checkpoint of inflammation.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Animales , Humanos , Ratones , Tejido Adiposo/metabolismo , Dieta Alta en Grasa/efectos adversos , Inflamasomas/genética , Inflamasomas/metabolismo , Inflamación/metabolismo , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Obesidad/metabolismo , Osteonectina/genética , Osteonectina/metabolismo
3.
Semin Immunol ; 70: 101818, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37611324

RESUMEN

T cells are a critical component of the immune system, found in abundance in blood, secondary lymphoid organs, and peripheral tissues. As individuals age, T cells are particularly susceptible to changes, making them one of the most affected immune subsets. These changes can have significant implications for age-related dysregulations, including the development of low-grade inflammation - a hallmark of aging known as inflammaging. In this review, we first present age-related changes in the functionality of the T cell compartment, including dysregulation of cytokine and chemokine production and cytotoxicity. Next, we discuss how these changes can contribute to the development and maintenance of inflammaging. Furthermore, we will summarize the mechanisms through which age-related changes in T cells may drive abnormal physiological outcomes.


Asunto(s)
Inflamación , Linfocitos T , Humanos , Envejecimiento , Citocinas
5.
Nature ; 615(7953): 668-677, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36890231

RESUMEN

Extracellular deposition of amyloid-ß as neuritic plaques and intracellular accumulation of hyperphosphorylated, aggregated tau as neurofibrillary tangles are two of the characteristic hallmarks of Alzheimer's disease1,2. The regional progression of brain atrophy in Alzheimer's disease highly correlates with tau accumulation but not amyloid deposition3-5, and the mechanisms of tau-mediated neurodegeneration remain elusive. Innate immune responses represent a common pathway for the initiation and progression of some neurodegenerative diseases. So far, little is known about the extent or role of the adaptive immune response and its interaction with the innate immune response in the presence of amyloid-ß or tau pathology6. Here we systematically compared the immunological milieux in the brain of mice with amyloid deposition or tau aggregation and neurodegeneration. We found that mice with tauopathy but not those with amyloid deposition developed a unique innate and adaptive immune response and that depletion of microglia or T cells blocked tau-mediated neurodegeneration. Numbers of T cells, especially those of cytotoxic T cells, were markedly increased in areas with tau pathology in mice with tauopathy and in the Alzheimer's disease brain. T cell numbers correlated with the extent of neuronal loss, and the cells dynamically transformed their cellular characteristics from activated to exhausted states along with unique TCR clonal expansion. Inhibition of interferon-γ and PDCD1 signalling both significantly ameliorated brain atrophy. Our results thus reveal a tauopathy- and neurodegeneration-related immune hub involving activated microglia and T cell responses, which could serve as therapeutic targets for preventing neurodegeneration in Alzheimer's disease and primary tauopathies.


Asunto(s)
Encéfalo , Microglía , Ovillos Neurofibrilares , Linfocitos T , Tauopatías , Animales , Ratones , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/inmunología , Péptidos beta-Amiloides/metabolismo , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Microglía/inmunología , Microglía/metabolismo , Ovillos Neurofibrilares/inmunología , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Proteínas tau/inmunología , Proteínas tau/metabolismo , Tauopatías/inmunología , Tauopatías/metabolismo , Tauopatías/patología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología , Placa Amiloide/inmunología , Placa Amiloide/metabolismo , Placa Amiloide/patología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/patología , Células Clonales/inmunología , Células Clonales/metabolismo , Células Clonales/patología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Inmunidad Innata
6.
Nat Cardiovasc Res ; 1(3): 263-280, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35959412

RESUMEN

Heart failure represents a major cause of morbidity and mortality worldwide. Single-cell transcriptomics have revolutionized our understanding of cell composition and associated gene expression. Through integrated analysis of single-cell and single-nucleus RNA-sequencing data generated from 27 healthy donors and 18 individuals with dilated cardiomyopathy, here we define the cell composition of the healthy and failing human heart. We identify cell-specific transcriptional signatures associated with age and heart failure and reveal the emergence of disease-associated cell states. Notably, cardiomyocytes converge toward common disease-associated cell states, whereas fibroblasts and myeloid cells undergo dramatic diversification. Endothelial cells and pericytes display global transcriptional shifts without changes in cell complexity. Collectively, our findings provide a comprehensive analysis of the cellular and transcriptomic landscape of human heart failure, identify cell type-specific transcriptional programs and disease-associated cell states and establish a valuable resource for the investigation of human heart failure.

7.
Nat Rev Immunol ; 22(8): 484-498, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34815556

RESUMEN

Ageing leads to profound alterations in the immune system and increases susceptibility to some chronic, infectious and autoimmune diseases. In recent years, widespread application of single-cell techniques has enabled substantial progress in our understanding of the ageing immune system. These comprehensive approaches have expanded and detailed the current views of ageing and immunity. Here we review a body of recent studies that explored how the immune system ages using unbiased profiling techniques at single-cell resolution. Specifically, we discuss an emergent understanding of age-related alterations in innate and adaptive immune cell populations, antigen receptor repertoires and immune cell-supporting microenvironments of the peripheral tissues. Focusing on the results obtained in mice and humans, we describe the multidimensional data that align with established concepts of immune ageing as well as novel insights emerging from these studies. We further discuss outstanding questions in the field and highlight techniques that will advance our understanding of immune ageing in the future.


Asunto(s)
Envejecimiento , Enfermedades Autoinmunes , Animales , Humanos , Sistema Inmunológico , Inmunidad Innata , Ratones
8.
Nat Aging ; 1(1): 124-141, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-34796338

RESUMEN

The impact of healthy aging on molecular programming of immune cells is poorly understood. Here, we report comprehensive characterization of healthy aging in human classical monocytes, with a focus on epigenomic, transcriptomic, and proteomic alterations, as well as the corresponding proteomic and metabolomic data for plasma, using healthy cohorts of 20 young and 20 older males (~27 and ~64 years old on average). For each individual, we performed eRRBS-based DNA methylation profiling, which allowed us to identify a set of age-associated differentially methylated regions (DMRs) - a novel, cell-type specific signature of aging in DNA methylome. Hypermethylation events were associated with H3K27me3 in the CpG islands near promoters of lowly-expressed genes, while hypomethylated DMRs were enriched in H3K4me1 marked regions and associated with age-related increase of expression of the corresponding genes, providing a link between DNA methylation and age-associated transcriptional changes in primary human cells.


Asunto(s)
Epigénesis Genética , Envejecimiento Saludable , Masculino , Humanos , Persona de Mediana Edad , Epigenoma , Monocitos , Proteómica , Metilación de ADN/genética
9.
Cell Metab ; 33(11): 2277-2287.e5, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34473956

RESUMEN

Aging impairs the integrated immunometabolic responses, which have evolved to maintain core body temperature in homeotherms to survive cold stress, infections, and dietary restriction. Adipose tissue inflammation regulates the thermogenic stress response, but how adipose tissue-resident cells instigate thermogenic failure in the aged are unknown. Here, we define alterations in the adipose-resident immune system and identify that type 2 innate lymphoid cells (ILC2s) are lost in aging. Restoration of ILC2 numbers in aged mice to levels seen in adults through IL-33 supplementation failed to rescue old mice from metabolic impairment and increased cold-induced lethality. Transcriptomic analyses revealed intrinsic defects in aged ILC2, and adoptive transfer of adult ILC2s are sufficient to protect old mice against cold. Thus, the functional defects in adipose ILC2s during aging drive thermogenic failure.


Asunto(s)
Inmunidad Innata , Interleucina-33 , Tejido Adiposo , Envejecimiento , Animales , Pulmón , Linfocitos , Ratones , Ratones Endogámicos C57BL
10.
Science ; 373(6553)2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34083450

RESUMEN

The meninges contain adaptive immune cells that provide immunosurveillance of the central nervous system (CNS). These cells are thought to derive from the systemic circulation. Through single-cell analyses, confocal imaging, bone marrow chimeras, and parabiosis experiments, we show that meningeal B cells derive locally from the calvaria, which harbors a bone marrow niche for hematopoiesis. B cells reach the meninges from the calvaria through specialized vascular connections. This calvarial-meningeal path of B cell development may provide the CNS with a constant supply of B cells educated by CNS antigens. Conversely, we show that a subset of antigen-experienced B cells that populate the meninges in aging mice are blood-borne. These results identify a private source for meningeal B cells, which may help maintain immune privilege within the CNS.


Asunto(s)
Subgrupos de Linfocitos B/fisiología , Linfocitos B/fisiología , Células de la Médula Ósea/fisiología , Sistema Nervioso Central/inmunología , Duramadre/citología , Linfopoyesis , Meninges/citología , Meninges/inmunología , Cráneo/anatomía & histología , Envejecimiento , Animales , Subgrupos de Linfocitos B/inmunología , Movimiento Celular , Sistema Nervioso Central/fisiología , Duramadre/inmunología , Fibroblastos/fisiología , Homeostasis , Privilegio Inmunológico , Ratones , Células Plasmáticas/fisiología , Análisis de la Célula Individual
11.
Elife ; 102021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34151773

RESUMEN

Increasing age is the strongest predictor of risk of COVID-19 severity and mortality. Immunometabolic switch from glycolysis to ketolysis protects against inflammatory damage and influenza infection in adults. To investigate how age compromises defense against coronavirus infection, and whether a pro-longevity ketogenic diet (KD) impacts immune surveillance, we developed an aging model of natural murine beta coronavirus (mCoV) infection with mouse hepatitis virus strain-A59 (MHV-A59). When inoculated intranasally, mCoV is pneumotropic and recapitulates several clinical hallmarks of COVID-19 infection. Aged mCoV-A59-infected mice have increased mortality and higher systemic inflammation in the heart, adipose tissue, and hypothalamus, including neutrophilia and loss of γδ T cells in lungs. Activation of ketogenesis in aged mice expands tissue protective γδ T cells, deactivates the NLRP3 inflammasome, and decreases pathogenic monocytes in lungs of infected aged mice. These data establish harnessing of the ketogenic immunometabolic checkpoint as a potential treatment against coronavirus infection in the aged.


Asunto(s)
Infecciones por Coronavirus/dietoterapia , Dieta Cetogénica/métodos , Virus de la Hepatitis Murina/patogenicidad , Factores de Edad , Envejecimiento , Animales , COVID-19/dietoterapia , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/mortalidad , Modelos Animales de Enfermedad , Glucólisis , Humanos , Inflamasomas/metabolismo , Cuerpos Cetónicos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Virus de la Hepatitis Murina/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , SARS-CoV-2
12.
Immunity ; 54(1): 99-115.e12, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33271118

RESUMEN

Systematic understanding of immune aging on a whole-body scale is currently lacking. We characterized age-associated alterations in immune cells across multiple mouse organs using single-cell RNA and antigen receptor sequencing and flow cytometry-based validation. We defined organ-specific and common immune alterations and identified a subpopulation of age-associated granzyme K (GZMK)-expressing CD8+ T (Taa) cells that are distinct from T effector memory (Tem) cells. Taa cells were highly clonal, had specific epigenetic and transcriptional signatures, developed in response to an aged host environment, and expressed markers of exhaustion and tissue homing. Activated Taa cells were the primary source of GZMK, which enhanced inflammatory functions of non-immune cells. In humans, proportions of the circulating GZMK+CD8+ T cell population that shares transcriptional and epigenetic signatures with mouse Taa cells increased during healthy aging. These results identify GZMK+ Taa cells as a potential target to address age-associated dysfunctions of the immune system.


Asunto(s)
Envejecimiento/fisiología , Linfocitos T CD8-positivos/fisiología , Sistema Inmunológico/fisiología , Inflamación/inmunología , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos T/genética , Animales , Células Cultivadas , Células Clonales , Citotoxicidad Inmunológica , Femenino , Perfilación de la Expresión Génica , Granzimas/metabolismo , Humanos , Memoria Inmunológica , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Transcriptoma
13.
bioRxiv ; 2020 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-33236006

RESUMEN

Increasing age is the strongest predictor of risk of COVID-19 severity. Unregulated cytokine storm together with impaired immunometabolic response leads to highest mortality in elderly infected with SARS-CoV-2. To investigate how aging compromises defense against COVID-19, we developed a model of natural murine beta coronavirus (mCoV) infection with mouse hepatitis virus strain MHV-A59 (mCoV-A59) that recapitulated majority of clinical hallmarks of COVID-19. Aged mCoV-A59-infected mice have increased mortality and higher systemic inflammation in the heart, adipose tissue and hypothalamus, including neutrophilia and loss of γδ T cells in lungs. Ketogenic diet increases beta-hydroxybutyrate, expands tissue protective γδ T cells, deactivates the inflammasome and decreases pathogenic monocytes in lungs of infected aged mice. These data underscore the value of mCoV-A59 model to test mechanism and establishes harnessing of the ketogenic immunometabolic checkpoint as a potential treatment against COVID-19 in the elderly. HIGHLIGHTS: - Natural MHV-A59 mouse coronavirus infection mimics COVID-19 in elderly.- Aged infected mice have systemic inflammation and inflammasome activation.- Murine beta coronavirus (mCoV) infection results in loss of pulmonary γδ T cells.- Ketones protect aged mice from infection by reducing inflammation. ETOC BLURB: Elderly have the greatest risk of death from COVID-19. Here, Ryu et al report an aging mouse model of coronavirus infection that recapitulates clinical hallmarks of COVID-19 seen in elderly. The increased severity of infection in aged animals involved increased inflammasome activation and loss of γδ T cells that was corrected by ketogenic diet.

14.
Nat Metab ; 2(1): 50-61, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-32694683

RESUMEN

Ketone bodies are essential alternative fuels that allow humans to survive periods of glucose scarcity induced by starvation and prolonged exercise. A widely used ketogenic diet (KD), which is extremely high in fat with very low carbohydrates, drives the host into using ß-hydroxybutyrate for the production of ATP and lowers NLRP3-mediated inflammation. However, the extremely high fat composition of KD raises the question of how ketogenesis affects adipose tissue to control inflammation and energy homeostasis. Here, by using single-cell RNA sequencing of adipose-tissue-resident immune cells, we show that KD expands metabolically protective γδ T cells that restrain inflammation. Notably, long-term ad libitum KD feeding in mice causes obesity, impairs metabolic health and depletes the adipose-resident γδ T cells. In addition, mice lacking γδ T cells have impaired glucose homeostasis. Our results suggest that γδ T cells are mediators of protective immunometabolic responses that link fatty acid-driven fuel use to reduced adipose tissue inflammation.


Asunto(s)
Grasa Intraabdominal/metabolismo , Cuerpos Cetónicos/biosíntesis , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/metabolismo , Ácido 3-Hidroxibutírico/metabolismo , Animales , Glucemia/metabolismo , Dieta Cetogénica , Homeostasis , Inflamación/metabolismo , Grasa Intraabdominal/inmunología , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Linfocitos T/inmunología
15.
Artículo en Inglés | MEDLINE | ID: mdl-32371549

RESUMEN

OBJECTIVE: To identify and characterize myeloid cell populations within the CSF of patients with MS and anti-myelin oligodendrocyte glycoprotein (MOG) disorder by high-resolution single-cell gene expression analysis. METHODS: Single-cell RNA sequencing (scRNA-seq) was used to profile individual cells of CSF and blood from 2 subjects with relapsing-remitting MS (RRMS) and one with anti-MOG disorder. Publicly available scRNA-seq data from the blood and CSF of 2 subjects with HIV were also analyzed. An informatics pipeline was used to cluster cell populations by transcriptomic profiling. Based on gene expression by CSF myeloid cells, a flow cytometry panel was devised to examine myeloid cell populations from the CSF of 11 additional subjects, including individuals with RRMS, anti-MOG disorder, and control subjects without inflammatory demyelination. RESULTS: Common myeloid populations were identified within the CSF of subjects with RRMS, anti-MOG disorder, and HIV. These included monocytes, conventional and plasmacytoid dendritic cells, and cells with a transcriptomic signature matching microglia. Microglia could be discriminated from other myeloid cell populations in the CSF by flow cytometry. CONCLUSIONS: High-resolution single-cell gene expression analysis clearly distinguishes distinct myeloid cell types present within the CSF of subjects with neuroinflammation. A population of microglia exists within the human CSF, which is detectable by surface protein expression. The function of these cells during immunity and disease requires further investigation.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso/líquido cefalorraquídeo , Líquido Cefalorraquídeo , Microglía , Células Mieloides , RNA-Seq , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Adulto , Anciano , Femenino , Infecciones por VIH/líquido cefalorraquídeo , Humanos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/líquido cefalorraquídeo , Glicoproteína Mielina-Oligodendrócito/inmunología , Adulto Joven
16.
Nat Immunol ; 20(6): 687-700, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31061528

RESUMEN

Most tissue-resident macrophage populations develop during embryogenesis, self-renew in the steady state and expand during type 2 immunity. Whether shared mechanisms regulate the proliferation of macrophages in homeostasis and disease is unclear. Here we found that the transcription factor Bhlhe40 was required in a cell-intrinsic manner for the self-renewal and maintenance of large peritoneal macrophages (LPMs), but not that of other tissue-resident macrophages. Bhlhe40 was necessary for the proliferation, but not the polarization, of LPMs in response to the cytokine IL-4. During infection with the helminth Heligmosomoides polygyrus bakeri, Bhlhe40 was required for cell cycling of LPMs. Bhlhe40 repressed the expression of genes encoding the transcription factors c-Maf and Mafb and directly promoted expression of transcripts encoding cell cycle-related proteins to enable the proliferation of LPMs. In LPMs, Bhlhe40 bound to genomic sites co-bound by the macrophage lineage-determining factor PU.1 and to unique sites, including Maf and loci encoding cell-cycle-related proteins. Our findings demonstrate a tissue-specific control mechanism that regulates the proliferation of resident macrophages in homeostasis and type 2 immunity.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Homeodominio/genética , Homeostasis/genética , Homeostasis/inmunología , Inmunidad/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Biomarcadores , Ciclo Celular/genética , Ciclo Celular/inmunología , Proliferación Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Proteínas de Homeodominio/metabolismo , Inmunofenotipificación , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Transgénicos , Monocitos/inmunología , Monocitos/metabolismo , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Transcriptoma
17.
J Exp Med ; 215(7): 1823-1838, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29773644

RESUMEN

The cytokine IL-10 antagonizes pathways that control Mycobacterium tuberculosis (Mtb) infection. Nevertheless, the impact of IL-10 during Mtb infection has been difficult to decipher because loss-of-function studies in animal models have yielded only mild phenotypes. We have discovered that the transcription factor basic helix-loop-helix family member e40 (Bhlhe40) is required to repress Il10 expression during Mtb infection. Loss of Bhlhe40 in mice results in higher Il10 expression, higher bacterial burden, and early susceptibility similar to that observed in mice lacking IFN-γ. Deletion of Il10 in Bhlhe40-/- mice reverses these phenotypes. Bhlhe40 deletion in T cells or CD11c+ cells is sufficient to cause susceptibility to Mtb Bhlhe40 represents the first transcription factor found to be essential during Mtb infection to specifically regulate Il10 expression, revealing the importance of strict control of IL-10 production by innate and adaptive immune cells during infection. Our findings uncover a previously elusive but significant role for IL-10 in Mtb pathogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Homeodominio/metabolismo , Interleucina-10/metabolismo , Proteínas Represoras/metabolismo , Tuberculosis/inmunología , Inmunidad Adaptativa , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Sitios Genéticos , Inmunidad Innata , Inflamación/patología , Interleucina-10/deficiencia , Linfocitos/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Células Mieloides/metabolismo , Neutrófilos/metabolismo , Unión Proteica , Células TH1/metabolismo , Tuberculosis/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA