Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Nat Immunol ; 17(6): 656-65, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27043409

RESUMEN

Group 2 innate lymphoid cells (ILC2s) regulate tissue inflammation and repair after activation by cell-extrinsic factors such as host-derived cytokines. However, the cell-intrinsic metabolic pathways that control ILC2 function are undefined. Here we demonstrate that expression of the enzyme arginase-1 (Arg1) during acute or chronic lung inflammation is a conserved trait of mouse and human ILC2s. Deletion of mouse ILC-intrinsic Arg1 abrogated type 2 lung inflammation by restraining ILC2 proliferation and dampening cytokine production. Mechanistically, inhibition of Arg1 enzymatic activity disrupted multiple components of ILC2 metabolic programming by altering arginine catabolism, impairing polyamine biosynthesis and reducing aerobic glycolysis. These data identify Arg1 as a key regulator of ILC2 bioenergetics that controls proliferative capacity and proinflammatory functions promoting type 2 inflammation.


Asunto(s)
Arginasa/metabolismo , Linfocitos/fisiología , Neumonía/inmunología , Animales , Arginasa/genética , Proliferación Celular/genética , Células Cultivadas , Citocinas/metabolismo , Glucólisis/genética , Humanos , Inmunidad Innata , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Poliaminas/metabolismo , Células Th2/inmunología
2.
Immunity ; 50(2): 505-519.e4, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30770247

RESUMEN

Innate lymphoid cells (ILC) play critical roles in regulating immunity, inflammation, and tissue homeostasis in mice. However, limited access to non-diseased human tissues has hindered efforts to profile anatomically-distinct ILCs in humans. Through flow cytometric and transcriptional analyses of lymphoid, mucosal, and metabolic tissues from previously healthy human organ donors, here we have provided a map of human ILC heterogeneity across multiple anatomical sites. In contrast to mice, human ILCs are less strictly compartmentalized and tissue localization selectively impacts ILC distribution in a subset-dependent manner. Tissue-specific distinctions are particularly apparent for ILC1 populations, whose distribution was markedly altered in obesity or aging. Furthermore, the degree of ILC1 population heterogeneity differed substantially in lymphoid versus mucosal sites. Together, these analyses comprise a comprehensive characterization of the spatial and temporal dynamics regulating the anatomical distribution, subset heterogeneity, and functional potential of ILCs in non-diseased human tissues.


Asunto(s)
Inmunidad Innata/inmunología , Linfocitos/inmunología , Especificidad de Órganos/inmunología , Transcriptoma/inmunología , Adolescente , Adulto , Anciano , Envejecimiento/genética , Animales , Niño , Preescolar , Femenino , Heterogeneidad Genética , Humanos , Inmunidad Innata/genética , Lactante , Recién Nacido , Linfocitos/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Especificidad de Órganos/genética , Transcriptoma/genética , Adulto Joven
3.
Nat Immunol ; 12(11): 1045-54, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21946417

RESUMEN

Innate lymphoid cells (ILCs), a heterogeneous cell population, are critical in orchestrating immunity and inflammation in the intestine, but whether ILCs influence immune responses or tissue homeostasis at other mucosal sites remains poorly characterized. Here we identify a population of lung-resident ILCs in mice and humans that expressed the alloantigen Thy-1 (CD90), interleukin 2 (IL-2) receptor a-chain (CD25), IL-7 receptor a-chain (CD127) and the IL-33 receptor subunit T1-ST2. Notably, mouse ILCs accumulated in the lung after infection with influenza virus, and depletion of ILCs resulted in loss of airway epithelial integrity, diminished lung function and impaired airway remodeling. These defects were restored by administration of the lung ILC product amphiregulin. Collectively, our results demonstrate a critical role for lung ILCs in restoring airway epithelial integrity and tissue homeostasis after infection with influenza virus.


Asunto(s)
Homeostasis , Inmunidad Innata , Gripe Humana/inmunología , Pulmón/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Orthomyxoviridae/inmunología , Mucosa Respiratoria/metabolismo , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Anfirregulina , Animales , Antígenos CD/biosíntesis , Células Cultivadas , Familia de Proteínas EGF , Glicoproteínas/farmacología , Homeostasis/inmunología , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Interleucina-33 , Interleucinas/metabolismo , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología , Cicatrización de Heridas
4.
Nature ; 549(7671): 282-286, 2017 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-28869965

RESUMEN

The type 2 cytokines interleukin (IL)-4, IL-5, IL-9 and IL-13 have important roles in stimulating innate and adaptive immune responses that are required for resistance to helminth infection, promotion of allergic inflammation, metabolic homeostasis and tissue repair. Group 2 innate lymphoid cells (ILC2s) produce type 2 cytokines, and although advances have been made in understanding the cytokine milieu that promotes ILC2 responses, how ILC2 responses are regulated by other stimuli remains poorly understood. Here we demonstrate that ILC2s in the mouse gastrointestinal tract co-localize with cholinergic neurons that express the neuropeptide neuromedin U (NMU). In contrast to other haematopoietic cells, ILC2s selectively express the NMU receptor 1 (NMUR1). In vitro stimulation of ILC2s with NMU induced rapid cell activation, proliferation, and secretion of the type 2 cytokines IL-5, IL-9 and IL-13 that was dependent on cell-intrinsic expression of NMUR1 and Gαq protein. In vivo administration of NMU triggered potent type 2 cytokine responses characterized by ILC2 activation, proliferation and eosinophil recruitment that was associated with accelerated expulsion of the gastrointestinal nematode Nippostrongylus brasiliensis or induction of lung inflammation. Conversely, worm burden was higher in Nmur1-/- mice than in control mice. Furthermore, use of gene-deficient mice and adoptive cell transfer experiments revealed that ILC2s were necessary and sufficient to mount NMU-elicited type 2 cytokine responses. Together, these data indicate that the NMU-NMUR1 neuronal signalling circuit provides a selective mechanism through which the enteric nervous system and innate immune system integrate to promote rapid type 2 cytokine responses that can induce anti-microbial, inflammatory and tissue-protective type 2 responses at mucosal sites.


Asunto(s)
Citocinas/inmunología , Inmunidad Innata , Inflamación/inmunología , Linfocitos/inmunología , Neuropéptidos/metabolismo , Traslado Adoptivo , Animales , Neuronas Colinérgicas/efectos de los fármacos , Neuronas Colinérgicas/metabolismo , Citocinas/metabolismo , Eosinófilos/citología , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/inervación , Inmunidad Innata/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/patología , Interleucina-13/inmunología , Interleucina-13/metabolismo , Interleucina-5/inmunología , Interleucina-5/metabolismo , Interleucina-9/inmunología , Interleucina-9/metabolismo , Linfocitos/citología , Linfocitos/efectos de los fármacos , Masculino , Ratones , Neuropéptidos/farmacología , Nippostrongylus/inmunología , Neumonía/inducido químicamente , Neumonía/inmunología , Neumonía/patología , Receptores de Neurotransmisores/deficiencia , Receptores de Neurotransmisores/genética , Receptores de Neurotransmisores/metabolismo , Transducción de Señal/efectos de los fármacos
5.
Immunity ; 38(4): 694-704, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23601684

RESUMEN

Group 2 innate lymphoid cells (ILC2) are innate lymphocytes that confer protective type 2 immunity during helminth infection and are also involved in allergic airway inflammation. Here we report that ILC2 development required T cell factor 1 (TCF-1, the product of the Tcf7 gene), a transcription factor also implicated in T cell lineage specification. Tcf7(-/-) mice lack ILC2, and were unable to mount ILC2-mediated innate type 2 immune responses. Forced expression of TCF-1 in bone marrow progenitors partially bypassed the requirement for Notch signaling in the generation of ILC2 in vivo. TCF-1 acted through both GATA-3-dependent and GATA-3-independent pathways to promote the generation of ILC2. These results are reminiscent of the critical roles of TCF-1 in early T cell development. Hence, transcription factors that underlie early steps of T cell development are also implicated in the development of innate lymphoid cells.


Asunto(s)
Asma/inmunología , Células de la Médula Ósea/inmunología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Linfocitos/inmunología , Nippostrongylus/inmunología , Infecciones por Strongylida/inmunología , Animales , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Factor Nuclear 1-alfa del Hepatocito/genética , Inmunidad Innata , Células Progenitoras Linfoides/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/genética , Transgenes/genética
6.
Immunol Rev ; 286(1): 137-147, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30294971

RESUMEN

Innate and adaptive lymphocytes employ diverse effector programs that provide optimal immunity to pathogens and orchestrate tissue homeostasis, or conversely can become dysregulated to drive progression of chronic inflammatory diseases. Emerging evidence suggests that CD4+ T helper cell subsets and their innate counterparts, the innate lymphoid cell family, accomplish these complex biological roles by selectively programming their cellular metabolism in order to instruct distinct modules of lymphocyte differentiation, proliferation, and cytokine production. Further, these metabolic pathways are significantly influenced by tissue microenvironments and disease states. Here, we summarize our current knowledge on how cell-intrinsic metabolic factors modulate the context-dependent bioenergetic pathways that govern innate and adaptive lymphocytes. Further, we propose that a greater understanding of these pathways may lead to the identification of unique features in each population and provoke the development of novel therapeutic strategies to modulate lymphocytes in health and disease.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Linfocitos/metabolismo , Animales , Diferenciación Celular , Microambiente Celular , Metabolismo Energético , Humanos , Inmunomodulación , Activación de Linfocitos , Linfocitos/inmunología
7.
Immunity ; 37(1): 158-70, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22705104

RESUMEN

Signals from commensal bacteria can influence immune cell development and susceptibility to infectious or inflammatory diseases. However, the mechanisms by which commensal bacteria regulate protective immunity after exposure to systemic pathogens remain poorly understood. Here, we demonstrate that antibiotic-treated (ABX) mice exhibit impaired innate and adaptive antiviral immune responses and substantially delayed viral clearance after exposure to systemic LCMV or mucosal influenza virus. Furthermore, ABX mice exhibited severe bronchiole epithelial degeneration and increased host mortality after influenza virus infection. Genome-wide transcriptional profiling of macrophages isolated from ABX mice revealed decreased expression of genes associated with antiviral immunity. Moreover, macrophages from ABX mice exhibited defective responses to type I and type II IFNs and impaired capacity to limit viral replication. Collectively, these data indicate that commensal-derived signals provide tonic immune stimulation that establishes the activation threshold of the innate immune system required for optimal antiviral immunity.


Asunto(s)
Bacterias/inmunología , Inmunidad Innata , Virus/inmunología , Inmunidad Adaptativa , Animales , Antibacterianos/farmacología , Infecciones por Arenaviridae/genética , Infecciones por Arenaviridae/inmunología , Bacterias/efectos de los fármacos , Susceptibilidad a Enfermedades/inmunología , Interferones/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología
8.
Nature ; 519(7542): 242-6, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25533952

RESUMEN

Obesity is an increasingly prevalent disease regulated by genetic and environmental factors. Emerging studies indicate that immune cells, including monocytes, granulocytes and lymphocytes, regulate metabolic homeostasis and are dysregulated in obesity. Group 2 innate lymphoid cells (ILC2s) can regulate adaptive immunity and eosinophil and alternatively activated macrophage responses, and were recently identified in murine white adipose tissue (WAT) where they may act to limit the development of obesity. However, ILC2s have not been identified in human adipose tissue, and the mechanisms by which ILC2s regulate metabolic homeostasis remain unknown. Here we identify ILC2s in human WAT and demonstrate that decreased ILC2 responses in WAT are a conserved characteristic of obesity in humans and mice. Interleukin (IL)-33 was found to be critical for the maintenance of ILC2s in WAT and in limiting adiposity in mice by increasing caloric expenditure. This was associated with recruitment of uncoupling protein 1 (UCP1)(+) beige adipocytes in WAT, a process known as beiging or browning that regulates caloric expenditure. IL-33-induced beiging was dependent on ILC2s, and IL-33 treatment or transfer of IL-33-elicited ILC2s was sufficient to drive beiging independently of the adaptive immune system, eosinophils or IL-4 receptor signalling. We found that ILC2s produce methionine-enkephalin peptides that can act directly on adipocytes to upregulate Ucp1 expression in vitro and that promote beiging in vivo. Collectively, these studies indicate that, in addition to responding to infection or tissue damage, ILC2s can regulate adipose function and metabolic homeostasis in part via production of enkephalin peptides that elicit beiging.


Asunto(s)
Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/inmunología , Inmunidad Innata/inmunología , Linfocitos/fisiología , Obesidad/inmunología , Adipocitos/citología , Adipocitos/efectos de los fármacos , Animales , Metabolismo Energético/inmunología , Encefalina Metionina/biosíntesis , Encefalina Metionina/metabolismo , Eosinófilos/inmunología , Eosinófilos/metabolismo , Femenino , Homeostasis/efectos de los fármacos , Humanos , Interleucinas/inmunología , Interleucinas/farmacología , Canales Iónicos/metabolismo , Linfocitos/citología , Linfocitos/inmunología , Masculino , Ratones , Proteínas Mitocondriales/metabolismo , Obesidad/patología , Receptores de Interleucina-4/inmunología , Receptores de Interleucina-4/metabolismo , Proteína Desacopladora 1
9.
Am J Respir Crit Care Med ; 201(1): 63-72, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31394048

RESUMEN

Rationale: Primary graft dysfunction (PGD) is the leading cause of early morbidity and mortality after lung transplantation, but the immunologic mechanisms are poorly understood. Innate lymphoid cells (ILC) are a heterogeneous family of immune cells regulating pathologic inflammation and beneficial tissue repair. However, whether changes in donor-derived lung ILC populations are associated with PGD development has never been examined.Objectives: To determine whether PGD in chronic obstructive pulmonary disease or interstitial lung disease transplant recipients is associated with alterations in ILC subset composition within the allograft.Methods: We performed a single-center cohort study of lung transplantation patients with surgical biopsies of donor tissue taken before, and immediately after, allograft reperfusion. Donor immune cells from 18 patients were characterized phenotypically by flow cytometry for single-cell resolution of distinct ILC subsets. Changes in the percentage of ILC subsets with reperfusion or PGD (grade 3 within 72 h) were assessed.Measurements and Main Results: Allograft reperfusion resulted in significantly decreased frequencies of natural killer cells and a trend toward reduced ILC populations, regardless of diagnosis (interstitial lung disease or chronic obstructive pulmonary disease). Seven patients developed PGD (38.9%), and PGD development was associated with selective reduction of the ILC2 subset after reperfusion. Conversely, patients without PGD exhibited significantly higher ILC1 frequencies before reperfusion, accompanied by elevated ILC2 frequencies after allograft reperfusion.Conclusions: The composition of donor ILC subsets is altered after allograft reperfusion and is associated with PGD development, suggesting that ILCs may be involved in regulating lung injury in lung transplant recipients.


Asunto(s)
Inmunidad Innata , Trasplante de Pulmón/efectos adversos , Linfocitos/inmunología , Disfunción Primaria del Injerto/etiología , Disfunción Primaria del Injerto/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factores de Riesgo
10.
Immunity ; 34(1): 122-34, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21194981

RESUMEN

Fetal CD4(+) lymphoid tissue inducer (LTi) cells play a critical role in the development of lymphoid tissues. Recent studies identified that LTi cells persist in adults and are related to a heterogeneous population of innate lymphoid cells that have been implicated in inflammatory responses. However, whether LTi cells contribute to protective immunity remains poorly defined. We demonstrate that after infection with Citrobacter rodentium, CD4(+) LTi cells were a dominant source of interleukin-22 (IL-22) early during infection. Infection-induced CD4(+) LTi cell responses were IL-23 dependent, and ablation of IL-23 impaired innate immunity. Further, depletion of CD4(+) LTi cells abrogated infection-induced expression of IL-22 and antimicrobial peptides, resulting in exacerbated host mortality. LTi cells were also found to be essential for host protective immunity in lymphocyte-replete hosts. Collectively these data demonstrate that adult CD4(+) LTi cells are a critical source of IL-22 and identify a previously unrecognized function for CD4(+) LTi cells in promoting innate immunity in the intestine.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Citrobacter rodentium/inmunología , Infecciones por Enterobacteriaceae/inmunología , Interleucina-23/metabolismo , Interleucinas/metabolismo , Intestinos/inmunología , Tejido Linfoide/patología , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Linfocitos T CD4-Positivos/patología , Células Cultivadas , Citrobacter rodentium/patogenicidad , Ensayo de Inmunoadsorción Enzimática , Inmunidad Innata , Inmunización , Interleucina-23/genética , Interleucina-23/inmunología , Interleucinas/genética , Interleucinas/inmunología , Intestinos/microbiología , Depleción Linfocítica , Tejido Linfoide/inmunología , Tejido Linfoide/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Interleucina-22
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA