Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cell ; 186(7): 1432-1447.e17, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-37001503

RESUMEN

Cancer immunotherapies, including adoptive T cell transfer, can be ineffective because tumors evolve to display antigen-loss-variant clones. Therapies that activate multiple branches of the immune system may eliminate escape variants. Here, we show that melanoma-specific CD4+ T cell therapy in combination with OX40 co-stimulation or CTLA-4 blockade can eradicate melanomas containing antigen escape variants. As expected, early on-target recognition of melanoma antigens by tumor-specific CD4+ T cells was required. Surprisingly, complete tumor eradication was dependent on neutrophils and partly dependent on inducible nitric oxide synthase. In support of these findings, extensive neutrophil activation was observed in mouse tumors and in biopsies of melanoma patients treated with immune checkpoint blockade. Transcriptomic and flow cytometry analyses revealed a distinct anti-tumorigenic neutrophil subset present in treated mice. Our findings uncover an interplay between T cells mediating the initial anti-tumor immune response and neutrophils mediating the destruction of tumor antigen loss variants.


Asunto(s)
Melanoma , Linfocitos T , Ratones , Animales , Linfocitos T/patología , Neutrófilos/patología , Deriva y Cambio Antigénico , Inmunoterapia , Antígeno CTLA-4
2.
Cell ; 184(19): 5015-5030.e16, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34407392

RESUMEN

Group 3 innate lymphoid cells (ILC3s) regulate immunity and inflammation, yet their role in cancer remains elusive. Here, we identify that colorectal cancer (CRC) manifests with altered ILC3s that are characterized by reduced frequencies, increased plasticity, and an imbalance with T cells. We evaluated the consequences of these changes in mice and determined that a dialog between ILC3s and T cells via major histocompatibility complex class II (MHCII) is necessary to support colonization with microbiota that subsequently induce type-1 immunity in the intestine and tumor microenvironment. As a result, mice lacking ILC3-specific MHCII develop invasive CRC and resistance to anti-PD-1 immunotherapy. Finally, humans with dysregulated intestinal ILC3s harbor microbiota that fail to induce type-1 immunity and immunotherapy responsiveness when transferred to mice. Collectively, these data define a protective role for ILC3s in cancer and indicate that their inherent disruption in CRC drives dysfunctional adaptive immunity, tumor progression, and immunotherapy resistance.


Asunto(s)
Neoplasias del Colon/inmunología , Neoplasias del Colon/terapia , Progresión de la Enfermedad , Inmunidad Innata , Inmunoterapia , Linfocitos/inmunología , Animales , Comunicación Celular/efectos de los fármacos , Plasticidad de la Célula/efectos de los fármacos , Neoplasias del Colon/microbiología , Heces/microbiología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunidad Innata/efectos de los fármacos , Inflamación/inmunología , Inflamación/patología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/patología , Intestinos/patología , Linfocitos/efectos de los fármacos , Ratones Endogámicos C57BL , Microbiota/efectos de los fármacos , Invasividad Neoplásica , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Donantes de Tejidos
3.
Immunity ; 52(4): 606-619.e6, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32160524

RESUMEN

Group 2 innate lymphoid cells (ILC2s) regulate immunity, inflammation, and tissue homeostasis. Two distinct subsets of ILC2s have been described: steady-state natural ILC2s and inflammatory ILC2s, which are elicited following helminth infection. However, how tissue-specific cues regulate these two subsets of ILC2s and their effector functions remains elusive. Here, we report that interleukin-33 (IL-33) promotes the generation of inflammatory ILC2s (ILC2INFLAM) via induction of the enzyme tryptophan hydroxylase 1 (Tph1). Tph1 expression was upregulated in ILC2s upon activation with IL-33 or following helminth infection in an IL-33-dependent manner. Conditional deletion of Tph1 in lymphocytes resulted in selective impairment of ILC2INFLAM responses and increased susceptibility to helminth infection. Further, RNA sequencing analysis revealed altered gene expression in Tph1 deficient ILC2s including inducible T cell co-stimulator (Icos). Collectively, these data reveal a previously unrecognized function for IL-33, Tph1, and ICOS in promoting inflammatory ILC2 responses and type 2 immunity at mucosal barriers.


Asunto(s)
Inmunidad Celular , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Interleucina-33/inmunología , Nippostrongylus/inmunología , Infecciones por Strongylida/inmunología , Subgrupos de Linfocitos T/inmunología , Triptófano Hidroxilasa/inmunología , Animales , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica/inmunología , Inmunidad Innata , Inmunidad Mucosa , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Interleucina-33/genética , Larva/crecimiento & desarrollo , Larva/inmunología , Larva/patogenicidad , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nippostrongylus/crecimiento & desarrollo , Nippostrongylus/patogenicidad , Cultivo Primario de Células , Transducción de Señal , Infecciones por Strongylida/genética , Infecciones por Strongylida/parasitología , Infecciones por Strongylida/patología , Subgrupos de Linfocitos T/clasificación , Subgrupos de Linfocitos T/parasitología , Triptófano Hidroxilasa/genética
4.
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
5.
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
6.
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
7.
J Immunol ; 193(5): 2416-26, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25057007

RESUMEN

Dendritic cells are major APCs that can efficiently prime immune responses. However, the roles of skin-resident Langerhans cells (LCs) in eliciting immune responses have not been fully understood. In this study, we demonstrate for the first time, to our knowledge, that LCs in cynomolgus macaque skin are capable of inducing antiviral-specific immune responses in vivo. Targeting HIV-Gag or influenza hemagglutinin Ags to skin LCs using recombinant fusion proteins of anti-Langerin Ab and Ags resulted in the induction of the viral Ag-specific responses. We further demonstrated that such Ag-specific immune responses elicited by skin LCs were greatly enhanced by TLR ligands, polyriboinosinic polyribocytidylic acid, and R848. These enhancements were not due to the direct actions of TLR ligands on LCs, but mainly dependent on TNF-α secreted from macrophages and neutrophils recruited to local tissues. Skin LC activation and migration out of the epidermis are associated with macrophage and neutrophil infiltration into the tissues. More importantly, blocking TNF-α abrogated the activation and migration of skin LCs. This study highlights that the cross-talk between innate immune cells in local tissues is an important component for the establishment of adaptive immunity. Understanding the importance of local immune networks will help us to design new and effective vaccines against microbial pathogens.


Asunto(s)
VIH-1/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/farmacología , Virus de la Influenza A/inmunología , Células de Langerhans/inmunología , Piel/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/farmacología , Inmunidad Adaptativa/efectos de los fármacos , Inmunidad Adaptativa/fisiología , Animales , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Imidazoles/farmacología , Macaca mulatta , Macrófagos/inmunología , Neutrófilos/inmunología , Poli I/farmacología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología
8.
Tetrahedron Lett ; 56(2): 458-460, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25601818

RESUMEN

Protein conjugates of toll-like receptor 7 agonists have been shown to elicit powerful immune responses. In order to facilitate our studies in this area our group has developed efficient syntheses for a number of functionalized derivatives that retain immune stimulatory activity.

9.
Semin Immunol ; 23(1): 21-7, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21277223

RESUMEN

Dendritic cells (DCs) were discovered in 1973 by Ralph Steinman as a previously undefined cell type in the mouse spleen and are now recognized as a group of related cell populations that induce and regulate adaptive immune responses. Studies of the past decade show that, both in mice and humans, DCs are composed of subsets that differ in their localization, phenotype, and functions. These progresses in our understanding of DC biology provide a new framework for improving human health. In this review, we discuss human DC subsets in the context of their medical applications, with a particular focus on DC targeting.


Asunto(s)
Células Dendríticas/inmunología , Vacunas/inmunología , Animales , Células Dendríticas/clasificación , Humanos
10.
J Immunol ; 189(5): 2645-55, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22865916

RESUMEN

Targeting of Ags directly to dendritic cells (DCs) through anti-DC receptor Ab fused to Ag proteins is a promising approach to vaccine development. However, not all Ags can be expressed as a rAb directly fused to a protein Ag. In this study, we show that noncovalent assembly of Ab-Ag complexes, mediated by interaction between dockerin and cohesin domains from cellulose-degrading bacteria, can greatly expand the range of Ags for this DC-targeting vaccine technology. rAbs with a dockerin domain fused to the rAb H chain C terminus are efficiently secreted by mammalian cells, and many Ags not secreted as rAb fusion proteins are readily expressed as cohesin directly fused to Ag either via secretion from mammalian cells or as soluble cytoplasmic Escherichia coli products. These form very stable and homogeneous complexes with rAb fused to dockerin. In vitro, these complexes can efficiently bind to human DC receptors followed by presentation to Ag-specific CD4⁺ and CD8⁺ T cells. Low doses of the HA1 subunit of influenza hemagglutinin conjugated through this means to anti-Langerin rAbs elicited Flu HA1-specific Ab and T cell responses in mice. Thus, the noncovalent assembly of rAb and Ag through dockerin and cohesin interaction provides a useful modular strategy for development and testing of prototype vaccines for elicitation of Ag-specific T and B cell responses, particularly when direct rAb fusions to Ag cannot be expressed.


Asunto(s)
Anticuerpos/administración & dosificación , Anticuerpos/fisiología , Antígenos/administración & dosificación , Antígenos/fisiología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Animales , Complejo Antígeno-Anticuerpo/administración & dosificación , Complejo Antígeno-Anticuerpo/biosíntesis , Complejo Antígeno-Anticuerpo/fisiología , Antígenos/aislamiento & purificación , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Diferenciación Celular/inmunología , Células Cultivadas , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Terapia Molecular Dirigida , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/fisiología
11.
Blood ; 116(10): 1685-97, 2010 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-20530286

RESUMEN

We evaluated human CD8(+) T-cell responses generated by targeting antigens to dendritic cells (DCs) through various lectin receptors. We found the immunoreceptor tyrosine-based inhibitory motif-containing DC immunoreceptor (DCIR) to mediate potent cross-presentation. A single exposure to a low dose of anti-DCIR-antigen conjugate initiated antigen-specific CD8(+) T-cell immunity by all human DC subsets including ex vivo-generated DCs, skin-isolated Langerhans cells, and blood myeloid DCs and plasmacytoid DCs. The delivery of influenza matrix protein (FluMP) through DCIR resulted in expansion of FluMP-specific memory CD8(+) T cells. Enhanced specific CD8(+) T-cell responses were observed when an antigen was delivered to the DCs via DCIR, compared with those induced by a free antigen, or antigen conjugated to a control monoclonal antibody or delivered via DC-SIGN, another lectin receptor. DCIR targeting also induced primary CD8(+) T-cell responses against self (MART-1) and viral (HIV gag) antigens. Addition of Toll-like receptor (TLR) 7/8 agonist enhanced DCIR-mediated cross-presentation as well as cross-priming, particularly when combined with a CD40 signal. TLR7/8 activation was associated with increased expansion of the primed CD8(+) T cells, high production of interferon-γ and tumor necrosis factor-α, and reduced levels of type 2-associated cytokines. Thus, antigen targeting via the human DCIR receptor allows activation of specific CD8(+) T-cell immunity.


Asunto(s)
Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Lectinas Tipo C/inmunología , Glicoproteínas de Membrana/inmunología , Receptores Inmunológicos/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Antígenos de Neoplasias/inmunología , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Reactividad Cruzada/efectos de los fármacos , Reactividad Cruzada/inmunología , Células Dendríticas/citología , Células Dendríticas/metabolismo , Citometría de Flujo , Humanos , Células de Langerhans/citología , Células de Langerhans/inmunología , Células de Langerhans/metabolismo , Lectinas Tipo C/metabolismo , Antígeno MART-1 , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Monocitos/citología , Monocitos/inmunología , Monocitos/metabolismo , Proteínas de Neoplasias/inmunología , Quinolinas/farmacología , Receptores Inmunológicos/metabolismo , Tiazoles/farmacología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología
12.
J Immunol ; 185(6): 3504-13, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20729328

RESUMEN

Dectin-1, a C-type lectin recognizing fungal and mycobacterial pathogens, can deliver intracellular signals that activate dendritic cells (DCs), resulting in initiation of immune responses and expansion of Th17 CD4(+) T cell responses. In this paper, we studied the roles of human Dectin-1 (hDectin-1) expressed on DCs in the induction and activation of Ag-specific CD8(+) T cell responses. We first generated an agonistic anti-hDectin-1 mAb, which recognizes the hDectin-1 Glu(143)-Ile(162) region. It bound to in vitro monocyte-derived DCs and to in vivo CD1c(+)CD1a(+) dermal DCs but not to epidermal Langerhans cells. Anti-hDectin-1-mediated DC activation resulted in upregulation of costimulatory molecules and secretion of multiple cytokines and chemokines in a Syk-dependent manner. DCs activated with the anti-hDectin-1 mAb could significantly enhance both neo and foreign Ag-specific CD8(+) T cell responses by promoting both the expansion of CD8(+) T cells and their functional activities. We further demonstrated that delivering Ags to DCs via hDectin-1 using anti-hDectin-1-Ag conjugates resulted in potent Ag-specific CD8(+) T cell responses. Thus, hDectin-1 expressed on DCs can contribute to the induction and activation of cellular immunity against intracellular pathogens, such as mycobacteria, that are recognized by DCs via Dectin-1. Vaccines based on delivering Ags to DCs with an agonistic anti-hDectin-1 mAb could elicit CD8(+) T cell-mediated immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/farmacología , Especificidad de Anticuerpos/genética , Sitios de Unión de Anticuerpos/genética , Línea Celular , Células Cultivadas , Epítopos de Linfocito T/genética , Humanos , Lectinas Tipo C , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/inmunología , Proteínas Recombinantes de Fusión/farmacología
13.
J Immunol ; 182(5): 2766-76, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19234171

RESUMEN

During viral infection, dendritic cells (DCs) capture infected cells and present viral Ags to CD8(+) T cells. However, activated DCs might potentially present cell-associated Ags derived from captured dead cells. In this study, we find that human DCs that captured dead cells containing the TLR3 agonist poly(I:C) produced cytokines and underwent maturation, but failed to elicit autologous CD8(+) T cell responses against Ags of dead cells. Accordingly, DCs that captured dead cells containing poly(I:C), or influenza virus, are unable to activate CD8(+) T cell clones specific to cell-associated Ags of captured dead cells. CD4(+) T cells are expanded with DCs that have captured poly(I:C)-containing dead cells, indicating the inhibition is specific for MHC class I-restricted cross-presentation. Furthermore, these DCs can expand naive allogeneic CD8(+) T cells. Finally, soluble or targeted Ag is presented when coloaded onto DCs that have captured poly(I:C)-containing dead cells, indicating the inhibition is specific for dead cell cargo that is accompanied by viral or poly(I:C) stimulus. Thus, DCs have a mechanism that prevents MHC class I-restricted cross-presentation of cell-associated Ag when they have captured dead infected cells.


Asunto(s)
Reactividad Cruzada/inmunología , Células Dendríticas/virología , Inhibidores de Crecimiento/inmunología , Antígeno HLA-A2/inmunología , Terapia de Inmunosupresión , Virus de la Influenza A/inmunología , Melanoma/virología , Poli I-C/inmunología , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Línea Celular Tumoral , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/inmunología , Células Dendríticas/patología , Antígeno HLA-A2/metabolismo , Humanos , Terapia de Inmunosupresión/métodos , Activación de Linfocitos/inmunología , Melanoma/inmunología , Melanoma/patología , Necrosis , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología , Linfocitos T Citotóxicos/virología
14.
Front Immunol ; 12: 735584, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34917073

RESUMEN

Common approaches for monitoring T cell responses are limited in their multiplexity and sensitivity. In contrast, deep sequencing of the T Cell Receptor (TCR) repertoire provides a global view that is limited only in terms of theoretical sensitivity due to the depth of available sampling; however, the assignment of antigen specificities within TCR repertoires has become a bottleneck. This study combines antigen-driven expansion, deep TCR sequencing, and a novel analysis framework to show that homologous 'Clusters of Expanded TCRs (CETs)' can be confidently identified without cell isolation, and assigned to antigen against a background of non-specific clones. We show that clonotypes within each CET respond to the same epitope, and that protein antigens stimulate multiple CETs reactive to constituent peptides. Finally, we demonstrate the personalized assignment of antigen-specificity to rare clones within fully-diverse uncultured repertoires. The method presented here may be used to monitor T cell responses to vaccination and immunotherapy with high fidelity.


Asunto(s)
Separación Celular/métodos , Técnicas Inmunológicas/métodos , Receptores de Antígenos de Linfocitos T/análisis , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Humanos
15.
Mucosal Immunol ; 13(4): 626-636, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32066836

RESUMEN

Thymic stromal lymphopoietin (TSLP) is a critical upstream cytokine inducing type 2 inflammation in various diseases, including asthma and atopic dermatitis. Accumulating evidence suggests that TSLP can directly stimulate a variety of immune cells, such as dendritic cells (DCs), basophils, T cells, and group 2 innate lymphoid cells (ILC2s). However, which cell types directly respond to TSLP in vivo and how TSLP initiates type 2 inflammation has remained controversial. To define the precise role of TSLP in vivo, for the first time we generated multiple cell lineage-specific TSLP receptor-deficient mice to systematically dissect the cell-intrinsic requirements for TSLP responsiveness in type 2 inflammation in the lung. In papain-induced innate immune-mediated type 2 airway inflammation, TSLP directly stimulated ILC2s, but not basophils, leading to enhanced type 2 inflammation. On the other hand, in OVA-induced adaptive immune-mediated type 2 airway inflammation, TSLP principally acted on DCs and CD4 + T cells during the sensitization phase, but not basophils or ILC2s, and facilitated the development of Th2 cell-mediated airway inflammation. Together, these findings reveal that TSLP activates distinct immune cell cascades in the context of innate and adaptive immune-mediated type 2 inflammation.


Asunto(s)
Susceptibilidad a Enfermedades , Técnicas de Silenciamiento del Gen , Inmunoglobulinas/genética , Inflamación/etiología , Inflamación/metabolismo , Receptores de Citocinas/genética , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Inmunidad Adaptativa , Animales , Biomarcadores , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Marcación de Gen , Inmunidad Innata , Inmunohistoquímica , Inflamación/patología , Ratones , Ratones Noqueados , Mucosa Respiratoria/patología
16.
J Exp Med ; 216(12): 2689-2700, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31601676

RESUMEN

Host-microbiota interactions are critical in regulating mammalian health and disease. In addition to bacteria, parasites, and viruses, beneficial communities of fungi (the mycobiome) are important modulators of immune- and tissue-homeostasis. Chitin is a major component of the fungal cell wall, and fibrinogen C containing domain 1 (FIBCD1) is a chitin-binding protein; however, the role of this molecule in influencing host-mycobiome interactions in vivo has never been examined. Here, we identify direct binding of FIBCD1 to intestinal-derived fungi and demonstrate that epithelial-specific expression of FIBCD1 results in significantly reduced fungal colonization and amelioration of fungal-driven intestinal inflammation. Collectively, these results identify FIBCD1 as a previously unrecognized microbial pattern recognition receptor through which intestinal epithelial cells can recognize and control fungal colonization, limit fungal dysbiosis, and dampen intestinal inflammation.


Asunto(s)
Hongos/fisiología , Interacciones Microbianas , Micobioma , Receptores de Superficie Celular/metabolismo , Animales , Quitina/metabolismo , ADN Espaciador Ribosómico , Modelos Animales de Enfermedad , Enteritis/etiología , Enteritis/metabolismo , Enteritis/patología , Microbioma Gastrointestinal , Expresión Génica , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Metagenómica , Ratones , Ratones Transgénicos , Unión Proteica , ARN Ribosómico 16S
17.
J Exp Med ; 216(12): 2714-2723, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31582416

RESUMEN

Host immunity to parasitic nematodes requires the generation of a robust type 2 cytokine response, characterized by the production of interleukin 13 (IL-13), which drives expulsion. Here, we show that infection with helminths in the intestine also induces an ILC2-driven, IL-13-dependent goblet cell hyperplasia and increased production of mucins (Muc5b and Muc5ac) at distal sites, including the lungs and other mucosal barrier sites. Critically, we show that type 2 priming of lung tissue through increased mucin production inhibits the progression of a subsequent lung migratory helminth infection and limits its transit through the airways. These data show that infection by gastrointestinal-dwelling helminths induces a systemic innate mucin response that primes peripheral barrier sites for protection against subsequent secondary helminth infections. These data suggest that innate-driven priming of mucus barriers may have evolved to protect from subsequent infections with multiple helminth species, which occur naturally in endemic areas.


Asunto(s)
Inmunidad Innata , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Moco/metabolismo , Animales , Protección Cruzada/inmunología , Células Caliciformes/citología , Células Caliciformes/metabolismo , Hiperplasia , Interleucina-13/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/parasitología , Masculino , Ratones , Ratones Noqueados , Mucinas/biosíntesis , Trichinella spiralis/inmunología , Triquinelosis/inmunología , Triquinelosis/parasitología
18.
Cell Rep ; 23(13): 3750-3758, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29949760

RESUMEN

The intestinal tract is constantly exposed to various stimuli. Group 3 innate lymphoid cells (ILC3s) reside in lymphoid organs and in the intestinal tract and are required for immunity to enteric bacterial infection. However, the mechanisms that regulate the ILC3s in vivo remain incompletely defined. Here, we show that GPR183, a chemotactic receptor expressed on murine and human ILC3s, regulates ILC3 migration toward its ligand 7α,25-dihydroxycholesterol (7α,25-OHC) in vitro, and GPR183 deficiency in vivo leads to a disorganized distribution of ILC3s in mesenteric lymph nodes and decreased ILC3 accumulation in the intestine. GPR183 functions intrinsically in ILC3s, and GPR183-deficient mice are more susceptible to enteric bacterial infection. Together, these results reveal a role for the GPR183-7α,25-OHC pathway in regulating the accumulation, distribution, and anti-microbial and tissue-protective functions of ILC3s and define a critical role for this pathway in promoting innate immunity to enteric bacterial infection.


Asunto(s)
Tejido Linfoide/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Movimiento Celular , Citrobacter rodentium/patogenicidad , Familia 7 del Citocromo P450/metabolismo , Infecciones por Enterobacteriaceae/patología , Infecciones por Enterobacteriaceae/prevención & control , Infecciones por Enterobacteriaceae/veterinaria , Humanos , Hidroxicolesteroles/química , Hidroxicolesteroles/metabolismo , Inmunidad Innata , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Ligandos , Tejido Linfoide/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Membrana Mucosa/citología , Membrana Mucosa/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Esteroide Hidroxilasas/deficiencia , Esteroide Hidroxilasas/genética , Esteroide Hidroxilasas/metabolismo
19.
PLoS One ; 13(11): e0207794, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30500852

RESUMEN

HIV-1 infection can be controlled by anti-retroviral drug therapy, but this is a lifetime treatment and the virus remains latent and rapidly rebounds if therapy is stopped. HIV-1-infected individuals under this drug regimen have increased rates of cancers, cardiovascular diseases, and autoimmunity due to compromised immunity. A therapeutic vaccine boosting cellular immunity against HIV-1 is therefore desirable and, possibly combined with other immune modulating agents, could obviate the need for long-term drug therapies. An approach to elicit strong T cell-based immunity is to direct virus protein antigens specifically to dendritic cells (DCs), which are the key cell type for controlling immune responses. For eliciting therapeutic cellular immunity in HIV-1-infected individuals, we developed vaccines comprised of five T cell epitope-rich regions of HIV-1 Gag, Nef, and Pol (HIV5pep) fused to monoclonal antibodies that bind either, the antigen presenting cell activating receptor CD40, or the endocytic dendritic cell immunoreceptor DCIR. The study aimed to demonstrate vaccine safety, establish efficacy for broad T cell responses in both primed and naïve settings, and identify one candidate vaccine for human therapeutic development. The vaccines were administered to Rhesus macaques by intradermal injection with poly-ICLC adjuvant. The animals were either i) naïve or, ii) previously primed with modified vaccinia Ankara vector (MVA) encoding HIV-1 Gag, Pol, and Nef (MVA GagPolNef). In the MVA-primed groups, both DC-targeting vaccinations boosted HIV5pep-specific blood CD4+ T cells producing multiple cytokines, but did not affect the MVA-elicited CD8+ T cell responses. In the naive groups, both DC-targeting vaccines elicited antigen-specific polyfunctional CD4+ and CD8+ T cell responses to multiple epitopes and these responses were unchanged by a subsequent MVA GagPolNef boost. In both settings, the T cell responses elicited via the CD40-targeting vaccine were more robust and were detectable in all the animals, favoring further development of the CD40-targeting vaccine for therapeutic vaccination of HIV-1-infected individuals.


Asunto(s)
Vacunas contra el SIDA/inmunología , Antígenos CD40/inmunología , Células Dendríticas/inmunología , Epítopos de Linfocito T/inmunología , Anticuerpos Anti-VIH/inmunología , VIH-1/inmunología , Receptores Inmunológicos/inmunología , Animales , Macaca mulatta , Masculino , Terapia Molecular Dirigida
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA