Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
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
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.
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
4.
Science ; 359(6379): 1056-1061, 2018 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-29496881

RESUMEN

The type 2 inflammatory response is induced by various environmental and infectious stimuli. Although recent studies identified group 2 innate lymphoid cells (ILC2s) as potent sources of type 2 cytokines, the molecular pathways controlling ILC2 responses are incompletely defined. Here we demonstrate that murine ILC2s express the ß2-adrenergic receptor (ß2AR) and colocalize with adrenergic neurons in the intestine. ß2AR deficiency resulted in exaggerated ILC2 responses and type 2 inflammation in intestinal and lung tissues. Conversely, ß2AR agonist treatment was associated with impaired ILC2 responses and reduced inflammation in vivo. Mechanistically, we demonstrate that the ß2AR pathway is a cell-intrinsic negative regulator of ILC2 responses through inhibition of cell proliferation and effector function. Collectively, these data provide the first evidence of a neuronal-derived regulatory circuit that limits ILC2-dependent type 2 inflammation.


Asunto(s)
Inmunidad Adaptativa , Neuronas Adrenérgicas/inmunología , Inmunidad Innata , Linfocitos/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Agonistas de Receptores Adrenérgicos beta 2/farmacología , Animales , Humanos , Inflamación/inmunología , Intestinos/inmunología , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Red Nerviosa/inmunología , Receptores Adrenérgicos beta 2/genética , Transducción de Señal
5.
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
6.
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
7.
Proc Natl Acad Sci U S A ; 112(34): 10762-7, 2015 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-26243875

RESUMEN

The barrier surfaces of the skin, lung, and intestine are constantly exposed to environmental stimuli that can result in inflammation and tissue damage. Interleukin (IL)-33-dependent group 2 innate lymphoid cells (ILC2s) are enriched at barrier surfaces and have been implicated in promoting inflammation; however, the mechanisms underlying the tissue-protective roles of IL-33 or ILC2s at surfaces such as the intestine remain poorly defined. Here we demonstrate that, following activation with IL-33, expression of the growth factor amphiregulin (AREG) is a dominant functional signature of gut-associated ILC2s. In the context of a murine model of intestinal damage and inflammation, the frequency and number of AREG-expressing ILC2s increases following intestinal injury and genetic disruption of the endogenous AREG-epidermal growth factor receptor (EGFR) pathway exacerbated disease. Administration of exogenous AREG limited intestinal inflammation and decreased disease severity in both lymphocyte-sufficient and lymphocyte-deficient mice, revealing a previously unrecognized innate immune mechanism of intestinal tissue protection. Furthermore, treatment with IL-33 or transfer of ILC2s ameliorated intestinal disease severity in an AREG-dependent manner. Collectively, these data reveal a critical feedback loop in which cytokine cues from damaged epithelia activate innate immune cells to express growth factors essential for ILC-dependent restoration of epithelial barrier function and maintenance of tissue homeostasis.


Asunto(s)
Colitis/inmunología , Familia de Proteínas EGF/fisiología , Receptores ErbB/fisiología , Inmunidad Innata/fisiología , Inmunidad Mucosa/fisiología , Interleucina-33/fisiología , Linfocitos/inmunología , Anfirregulina , Animales , Colitis/inducido químicamente , Colitis/terapia , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Familia de Proteínas EGF/deficiencia , Familia de Proteínas EGF/uso terapéutico , Epitelio/inmunología , Epitelio/metabolismo , Epitelio/patología , Retroalimentación Fisiológica , Inmunoterapia Adoptiva , Interleucina-33/biosíntesis , Interleucina-33/genética , Interleucina-33/uso terapéutico , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Pulmón/inmunología , Pulmón/patología , Linfocitos/clasificación , Ratones , Ratones Noqueados , Mucinas/biosíntesis , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas Recombinantes/uso terapéutico , Transducción de Señal , Organismos Libres de Patógenos Específicos
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.
Science ; 345(6196): 578-82, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25082704

RESUMEN

The mammalian intestine is colonized by beneficial commensal bacteria and is a site of infection by pathogens, including helminth parasites. Helminths induce potent immunomodulatory effects, but whether these effects are mediated by direct regulation of host immunity or indirectly through eliciting changes in the microbiota is unknown. We tested this in the context of virus-helminth coinfection. Helminth coinfection resulted in impaired antiviral immunity and was associated with changes in the microbiota and STAT6-dependent helminth-induced alternative activation of macrophages. Notably, helminth-induced impairment of antiviral immunity was evident in germ-free mice, but neutralization of Ym1, a chitinase-like molecule that is associated with alternatively activated macrophages, could partially restore antiviral immunity. These data indicate that helminth-induced immunomodulation occurs independently of changes in the microbiota but is dependent on Ym1.


Asunto(s)
Infecciones por Caliciviridae/inmunología , Coinfección/inmunología , Gastroenteritis/inmunología , Inmunomodulación , Lectinas/inmunología , Microbiota/inmunología , Norovirus/inmunología , Trichinella/inmunología , Triquinelosis/inmunología , beta-N-Acetilhexosaminidasas/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Coinfección/microbiología , Coinfección/parasitología , Gastroenteritis/virología , Vida Libre de Gérmenes , Intestinos/inmunología , Intestinos/microbiología , Intestinos/virología , Activación de Macrófagos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL
10.
J Immunol ; 193(7): 3717-25, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25156365

RESUMEN

Type 2 inflammation underlies allergic diseases such as atopic dermatitis, which is characterized by the accumulation of basophils and group 2 innate lymphoid cells (ILC2s) in inflamed skin lesions. Although murine studies have demonstrated that cutaneous basophil and ILC2 responses are dependent on thymic stromal lymphopoietin, whether these cell populations interact to regulate the development of cutaneous type 2 inflammation is poorly defined. In this study, we identify that basophils and ILC2s significantly accumulate in inflamed human and murine skin and form clusters not observed in control skin. We demonstrate that murine basophil responses precede ILC2 responses and that basophils are the dominant IL-4-enhanced GFP-expressing cell type in inflamed skin. Furthermore, basophils and IL-4 were necessary for the optimal accumulation of ILC2s and induction of atopic dermatitis-like disease. We show that ILC2s express IL-4Rα and proliferate in an IL-4-dependent manner. Additionally, basophil-derived IL-4 was required for cutaneous ILC2 responses in vivo and directly regulated ILC2 proliferation ex vivo. Collectively, these data reveal a previously unrecognized role for basophil-derived IL-4 in promoting ILC2 responses during cutaneous inflammation.


Asunto(s)
Basófilos/inmunología , Dermatitis Atópica/inmunología , Inmunidad Innata , Linfocitos/inmunología , Piel/inmunología , Animales , Basófilos/patología , Proliferación Celular , Citocinas/genética , Citocinas/inmunología , Dermatitis Atópica/genética , Dermatitis Atópica/patología , Femenino , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-4/genética , Interleucina-4/inmunología , Linfocitos/patología , Masculino , Ratones , Ratones Noqueados , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Piel/patología , Linfopoyetina del Estroma Tímico
11.
J Exp Med ; 210(9): 1823-37, 2013 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-23960191

RESUMEN

The predominantly epithelial cell-derived cytokines IL-25, IL-33, and thymic stromal lymphopoietin (TSLP) can promote CD4(+) Th2 cell-dependent immunity, inflammation, and tissue repair at barrier surfaces through the induction of multiple innate immune cell populations. IL-25 and IL-33 were previously shown to elicit four innate cell populations, named natural helper cells, nuocytes, innate type 2 helper cells, and multipotent progenitor type 2 (MPP(type2)) cells, now collectively termed group 2 innate lymphoid cells (ILC2). In contrast to other types of ILC2, MPP(type2) cells exhibit multipotent potential and do not express T1/ST2 or IL-7Rα, suggesting that MPP(type2) cells may be a distinct population. Here, we show that IL-33 elicits robust ILC2 responses, whereas IL-25 predominantly promotes MPP(type2) cell responses at multiple tissue sites with limited effects on ILC2 responses. MPP(type2) cells were distinguished from ILC2 by their differential developmental requirements for specific transcription factors, distinct genome-wide transcriptional profile, and functional potential. Furthermore, IL-25-induced MPP(type2) cells promoted Th2 cytokine-associated inflammation after depletion of ILC2. These findings indicate that IL-25 simultaneously elicits phenotypically and functionally distinct innate lymphoid- and nonlymphoid-associated cell populations and implicate IL-25-elicited MPP(type2) cells and extramedullary hematopoiesis in the promotion of Th2 cytokine responses at mucosal surfaces.


Asunto(s)
Inmunidad Innata/efectos de los fármacos , Interleucina-17/farmacología , Linfocitos/citología , Linfocitos/inmunología , Células Madre Multipotentes/citología , Células Madre Multipotentes/inmunología , Animales , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Inflamación/inmunología , Inflamación/patología , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Interleucina-33 , Interleucinas/metabolismo , Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Células Madre Multipotentes/efectos de los fármacos , Fenotipo , Unión Proteica/efectos de los fármacos , Unión Proteica/inmunología , Receptores de Interleucina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Células Th2/inmunología , Transcripción Genética/efectos de los fármacos , Transcriptoma
12.
Nature ; 498(7452): 113-7, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23698371

RESUMEN

Innate lymphoid cells (ILCs) are a recently characterized family of immune cells that have critical roles in cytokine-mediated regulation of intestinal epithelial cell barrier integrity. Alterations in ILC responses are associated with multiple chronic human diseases, including inflammatory bowel disease, implicating a role for ILCs in disease pathogenesis. Owing to an inability to target ILCs selectively, experimental studies assessing ILC function have predominantly used mice lacking adaptive immune cells. However, in lymphocyte-sufficient hosts ILCs are vastly outnumbered by CD4(+) T cells, which express similar profiles of effector cytokines. Therefore, the function of ILCs in the presence of adaptive immunity and their potential to influence adaptive immune cell responses remain unknown. To test this, we used genetic or antibody-mediated depletion strategies to target murine ILCs in the presence of an adaptive immune system. We show that loss of retinoic-acid-receptor-related orphan receptor-γt-positive (RORγt(+)) ILCs was associated with dysregulated adaptive immune cell responses against commensal bacteria and low-grade systemic inflammation. Remarkably, ILC-mediated regulation of adaptive immune cells occurred independently of interleukin (IL)-17A, IL-22 or IL-23. Genome-wide transcriptional profiling and functional analyses revealed that RORγt(+) ILCs express major histocompatibility complex class II (MHCII) and can process and present antigen. However, rather than inducing T-cell proliferation, ILCs acted to limit commensal bacteria-specific CD4(+) T-cell responses. Consistent with this, selective deletion of MHCII in murine RORγt(+) ILCs resulted in dysregulated commensal bacteria-dependent CD4(+) T-cell responses that promoted spontaneous intestinal inflammation. These data identify that ILCs maintain intestinal homeostasis through MHCII-dependent interactions with CD4(+) T cells that limit pathological adaptive immune cell responses to commensal bacteria.


Asunto(s)
Bacterias/inmunología , Linfocitos T CD4-Positivos/inmunología , Inmunidad Innata/inmunología , Intestinos/inmunología , Intestinos/microbiología , Simbiosis , Animales , Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/patología , Proliferación Celular , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Inflamación/patología , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Interleucinas/metabolismo , Intestinos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Interleucina-22
13.
J Virol ; 87(12): 7015-31, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23596300

RESUMEN

Norovirus (NV) gastroenteritis is a major contributor to global morbidity and mortality, yet little is known about immune mechanisms leading to NV control. Previous studies using the murine norovirus (MNV) model have established a key role for T cells in MNV clearance. Despite these advances, important questions remain regarding the magnitude, location, and dynamics of the MNV-specific T cell response. To address these questions, we identified MNV-specific major histocompatibility complex (MHC) class I immunodominant epitopes using an overlapping peptide screen. One of these epitopes (amino acids 519 to 527 of open reading frame 2 [ORF2(519-527)]) was highly conserved among all NV genogroups. Using MHC class I peptide tetramers, we tracked MNV-specific CD8 T cells in lymphoid and mucosal sites during infection with two MNV strains with distinct biological behaviors, the acutely cleared strain CW3 and the persistent strain CR6. Here, we show that enteric MNV infection elicited robust T cell responses primarily in the intestinal mucosa and that MNV-specific CD8 T cells dynamically regulated the expression of surface molecules associated with activation, differentiation, and homing. Furthermore, compared to MNV-CW3 infection, chronic infection with MNV-CR6 resulted in fewer and less-functional CD8 T cells, and this difference was evident as early as day 8 postinfection. Finally, MNV-specific CD8 T cells were capable of reducing the viral load in persistently infected Rag1(-/-) mice, suggesting that these cells are a crucial component of NV immunity. Collectively, these data provide fundamental new insights into the adaptive immune response to two closely related NV strains with distinct biological behaviors and bring us closer to understanding the correlates of protective antiviral immunity in the intestine.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Caliciviridae/inmunología , Gastroenteritis/inmunología , Mucosa Intestinal , Norovirus/inmunología , Animales , Infecciones por Caliciviridae/virología , Enfermedad Crónica , Epítopos de Linfocito T , Femenino , Citometría de Flujo , Gastroenteritis/virología , Epítopos Inmunodominantes , Mucosa Intestinal/inmunología , Mucosa Intestinal/virología , Activación de Linfocitos , Complejo Mayor de Histocompatibilidad , Ratones , Ratones Endogámicos C57BL
14.
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
15.
PLoS Pathog ; 9(3): e1003207, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23516357

RESUMEN

Seasonal epidemics of influenza virus result in ∼36,000 deaths annually in the United States. Current vaccines against influenza virus elicit an antibody response specific for the envelope glycoproteins. However, high mutation rates result in the emergence of new viral serotypes, which elude neutralization by preexisting antibodies. T lymphocytes have been reported to be capable of mediating heterosubtypic protection through recognition of internal, more conserved, influenza virus proteins. Here, we demonstrate using a recombinant influenza virus expressing the LCMV GP33-41 epitope that influenza virus-specific CD8+ T cells and virus-specific non-neutralizing antibodies each are relatively ineffective at conferring heterosubtypic protective immunity alone. However, when combined virus-specific CD8 T cells and non-neutralizing antibodies cooperatively elicit robust protective immunity. This synergistic improvement in protective immunity is dependent, at least in part, on alveolar macrophages and/or other lung phagocytes. Overall, our studies suggest that an influenza vaccine capable of eliciting both CD8+ T cells and antibodies specific for highly conserved influenza proteins may be able to provide heterosubtypic protection in humans, and act as the basis for a potential "universal" vaccine.


Asunto(s)
Anticuerpos Antivirales/inmunología , Linfocitos T CD8-positivos/inmunología , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Macrófagos Alveolares/inmunología , Inmunidad Adaptativa , Animales , Anticuerpos Neutralizantes/inmunología , Antígenos Virales/inmunología , Línea Celular , Protección Cruzada , Perros , Femenino , Glicoproteínas/inmunología , Humanos , Gripe Humana/inmunología , Gripe Humana/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fragmentos de Péptidos/inmunología , Carga Viral , Proteínas Virales/inmunología
16.
Sci Transl Med ; 5(170): 170ra16, 2013 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-23363980

RESUMEN

Innate lymphoid cells (ILCs) are a recently identified family of heterogeneous immune cells that can be divided into three groups based on their differential developmental requirements and expression of effector cytokines. Among these, group 2 ILCs produce the type 2 cytokines interleukin-5 (IL-5) and IL-13 and promote type 2 inflammation in the lung and intestine. However, whether group 2 ILCs reside in the skin and contribute to skin inflammation has not been characterized. We identify a population of skin-resident group 2 ILCs present in healthy human skin that are enriched in lesional human skin from atopic dermatitis (AD) patients. Group 2 ILCs were also found in normal murine skin and were critical for the development of inflammation in a murine model of AD-like disease. Remarkably, in contrast to group 2 ILC responses in the intestine and lung, which are critically regulated by IL-33 and IL-25, group 2 ILC responses in the skin and skin-draining lymph nodes were independent of these canonical cytokines but were critically dependent on thymic stromal lymphopoietin (TSLP). Collectively, these results demonstrate an essential role for IL-33- and IL-25-independent group 2 ILCs in promoting skin inflammation.


Asunto(s)
Citocinas/metabolismo , Inmunidad Innata/inmunología , Inflamación/inmunología , Interleucinas/metabolismo , Linfocitos/inmunología , Piel/patología , Inmunidad Adaptativa , Animales , Dermatitis Atópica/inmunología , Dermatitis Atópica/patología , Modelos Animales de Enfermedad , Humanos , Inflamación/patología , Interleucina-33 , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Piel/inmunología , Linfopoyetina del Estroma Tímico
17.
Science ; 336(6086): 1321-5, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22674331

RESUMEN

The mammalian intestinal tract is colonized by trillions of beneficial commensal bacteria that are anatomically restricted to specific niches. However, the mechanisms that regulate anatomical containment remain unclear. Here, we show that interleukin-22 (IL-22)-producing innate lymphoid cells (ILCs) are present in intestinal tissues of healthy mammals. Depletion of ILCs resulted in peripheral dissemination of commensal bacteria and systemic inflammation, which was prevented by administration of IL-22. Disseminating bacteria were identified as Alcaligenes species originating from host lymphoid tissues. Alcaligenes was sufficient to promote systemic inflammation after ILC depletion in mice, and Alcaligenes-specific systemic immune responses were associated with Crohn's disease and progressive hepatitis C virus infection in patients. Collectively, these data indicate that ILCs regulate selective containment of lymphoid-resident bacteria to prevent systemic inflammation associated with chronic diseases.


Asunto(s)
Alcaligenes/fisiología , Interleucinas/inmunología , Intestinos/inmunología , Linfocitos/inmunología , Tejido Linfoide/inmunología , Tejido Linfoide/microbiología , Adulto , Alcaligenes/inmunología , Alcaligenes/aislamiento & purificación , Animales , Traslocación Bacteriana , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/microbiología , Hepatitis C Crónica/inmunología , Hepatitis C Crónica/microbiología , Humanos , Inmunidad Innata , Inflamación , Interleucinas/administración & dosificación , Interleucinas/biosíntesis , Intestinos/microbiología , Complejo de Antígeno L1 de Leucocito/metabolismo , Hígado/microbiología , Ganglios Linfáticos/inmunología , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Bazo/microbiología , Adulto Joven , Interleucina-22
18.
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
19.
Curr Opin Immunol ; 24(3): 284-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22521139

RESUMEN

Maintenance of epithelial barrier function in the skin, respiratory tract and intestine is critical to limit exposure to commensal and pathogenic microbes and to maintain tissue homeostasis. Innate lymphoid cells (ILCs) are a recently recognized innate immune cell population that plays critical roles in host defense, regulation of inflammation and promotion of wound healing and tissue repair at barrier surfaces. In this review we discuss recent advances in the understanding of how ILC populations in the respiratory tract impact allergic airway inflammation and lung epithelial repair.


Asunto(s)
Asma/inmunología , Inmunidad Innata , Linfocitos/inmunología , Animales , Células Epiteliales/inmunología , Humanos
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...