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.
Artigo em Inglês | MEDLINE | ID: mdl-37001503

RESUMO

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.


Assuntos
Melanoma , Linfócitos T , Camundongos , Animais , Linfócitos T/patologia , Neutrófilos/patologia , Deriva e Deslocamento Antigênicos , Imunoterapia , Antígeno CTLA-4
2.
Cell ; 184(19): 5015-5030.e16, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34407392

RESUMO

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.


Assuntos
Neoplasias do Colo/imunologia , Neoplasias do Colo/terapia , Progressão da Doença , Imunidade Inata , Imunoterapia , Linfócitos/imunologia , Animais , Comunicação Celular/efeitos dos fármacos , Plasticidade Celular/efeitos dos fármacos , Neoplasias do Colo/microbiologia , Fezes/microbiologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Imunidade Inata/efeitos dos fármacos , Inflamação/imunologia , Inflamação/patologia , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/patologia , Intestinos/patologia , Linfócitos/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Microbiota/efeitos dos fármacos , Invasividade Neoplásica , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Doadores de Tecidos
3.
Immunity ; 52(4): 606-619.e6, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32160524

RESUMO

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.


Assuntos
Imunidade Celular , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Interleucina-33/imunologia , Nippostrongylus/imunologia , Infecções por Strongylida/imunologia , Subpopulações de Linfócitos T/imunologia , Triptofano Hidroxilase/imunologia , Animais , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Suscetibilidade a Doenças , Regulação da Expressão Gênica/imunologia , Imunidade Inata , Imunidade nas Mucosas , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Interleucina-33/genética , Larva/crescimento & desenvolvimento , Larva/imunologia , Larva/patogenicidade , Linfonodos/imunologia , Linfonodos/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nippostrongylus/crescimento & desenvolvimento , Nippostrongylus/patogenicidade , Cultura Primária de Células , Transdução de Sinais , Infecções por Strongylida/genética , Infecções por Strongylida/parasitologia , Infecções por Strongylida/patologia , Subpopulações de Linfócitos T/classificação , Subpopulações de Linfócitos T/parasitologia , Triptofano Hidroxilase/genética
4.
Nat Immunol ; 17(6): 656-65, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27043409

RESUMO

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.


Assuntos
Arginase/metabolismo , Linfócitos/fisiologia , Pneumonia/imunologia , Animais , Arginase/genética , Proliferação de Células/genética , Células Cultivadas , Citocinas/metabolismo , Glicólise/genética , Humanos , Imunidade Inata , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Poliaminas/metabolismo , Células Th2/imunologia
5.
Immunity ; 50(2): 505-519.e4, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30770247

RESUMO

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.


Assuntos
Imunidade Inata/imunologia , Linfócitos/imunologia , Especificidade de Órgãos/imunologia , Transcriptoma/imunologia , Adolescente , Adulto , Idoso , Envelhecimento/genética , Animais , Criança , Pré-Escolar , Feminino , Heterogeneidade Genética , Humanos , Imunidade Inata/genética , Lactente , Recém-Nascido , Linfócitos/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Especificidade de Órgãos/genética , Transcriptoma/genética , Adulto Jovem
6.
Nature ; 549(7671): 282-286, 2017 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-28869965

RESUMO

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.


Assuntos
Citocinas/imunologia , Imunidade Inata , Inflamação/imunologia , Linfócitos/imunologia , Neuropeptídeos/metabolismo , Transferência Adotiva , Animais , Neurônios Colinérgicos/efeitos dos fármacos , Neurônios Colinérgicos/metabolismo , Citocinas/metabolismo , Eosinófilos/citologia , Eosinófilos/efeitos dos fármacos , Eosinófilos/imunologia , Feminino , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Trato Gastrointestinal/citologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/inervação , Imunidade Inata/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/patologia , Interleucina-13/imunologia , Interleucina-13/metabolismo , Interleucina-5/imunologia , Interleucina-5/metabolismo , Interleucina-9/imunologia , Interleucina-9/metabolismo , Linfócitos/citologia , Linfócitos/efeitos dos fármacos , Masculino , Camundongos , Neuropeptídeos/farmacologia , Nippostrongylus/imunologia , Pneumonia/induzido quimicamente , Pneumonia/imunologia , Pneumonia/patologia , Receptores de Neurotransmissores/deficiência , Receptores de Neurotransmissores/genética , Receptores de Neurotransmissores/metabolismo , Transdução de Sinais/efeitos dos fármacos
7.
J Immunol ; 193(5): 2416-26, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25057007

RESUMO

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.


Assuntos
HIV-1/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/farmacologia , Vírus da Influenza A/imunologia , Células de Langerhans/imunologia , Pele/imunologia , Fator de Necrose Tumoral alfa/imunologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/farmacologia , Imunidade Adaptativa/efeitos dos fármacos , Imunidade Adaptativa/fisiologia , Animais , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Imidazóis/farmacologia , Macaca mulatta , Macrófagos/imunologia , Neutrófilos/imunologia , Poli I/farmacologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia
8.
Tetrahedron Lett ; 56(2): 458-460, 2015 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-25601818

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-21277223

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Vacinas/imunologia , Animais , Células Dendríticas/classificação , Humanos
10.
J Immunol ; 189(5): 2645-55, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22865916

RESUMO

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.


Assuntos
Anticorpos/administração & dosagem , Anticorpos/fisiologia , Antígenos/administração & dosagem , Antígenos/fisiologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Animais , Complexo Antígeno-Anticorpo/administração & dosagem , Complexo Antígeno-Anticorpo/biossíntese , Complexo Antígeno-Anticorpo/fisiologia , Antígenos/isolamento & purificação , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/imunologia , Células Cultivadas , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Terapia de Alvo Molecular , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/fisiologia
11.
Blood ; 116(10): 1685-97, 2010 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-20530286

RESUMO

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.


Assuntos
Antígenos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Lectinas Tipo C/imunologia , Glicoproteínas de Membrana/imunologia , Receptores Imunológicos/imunologia , Animais , Anticorpos Monoclonais/imunologia , Antígenos de Neoplasias/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Células Cultivadas , Apresentação Cruzada/efeitos dos fármacos , Apresentação Cruzada/imunologia , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Citometria de Fluxo , Humanos , Células de Langerhans/citologia , Células de Langerhans/imunologia , Células de Langerhans/metabolismo , Lectinas Tipo C/metabolismo , Antígeno MART-1 , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/citologia , Monócitos/imunologia , Monócitos/metabolismo , Proteínas de Neoplasias/imunologia , Quinolinas/farmacologia , Receptores Imunológicos/metabolismo , Tiazóis/farmacologia , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia
12.
J Immunol ; 185(6): 3504-13, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20729328

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Proteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacologia , Especificidade de Anticorpos/genética , Sítios de Ligação de Anticorpos/genética , Linhagem Celular , Células Cultivadas , Epitopos de Linfócito T/genética , Humanos , Lectinas Tipo C , Proteínas de Membrana/agonistas , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/agonistas , Proteínas do Tecido Nervoso/imunologia , Proteínas Recombinantes de Fusão/farmacologia
13.
J Immunol ; 182(5): 2766-76, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19234171

RESUMO

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.


Assuntos
Apresentação Cruzada/imunologia , Células Dendríticas/virologia , Inibidores do Crescimento/imunologia , Antígeno HLA-A2/imunologia , Terapia de Imunossupressão , Vírus da Influenza A/imunologia , Melanoma/virologia , Poli I-C/imunologia , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/imunologia , Células Dendríticas/patologia , Antígeno HLA-A2/metabolismo , Humanos , Terapia de Imunossupressão/métodos , Ativação Linfocitária/imunologia , Melanoma/imunologia , Melanoma/patologia , Necrose , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/patologia , Linfócitos T Citotóxicos/virologia
14.
Front Immunol ; 12: 735584, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34917073

RESUMO

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.


Assuntos
Separação Celular/métodos , Técnicas Imunológicas/métodos , Receptores de Antígenos de Linfócitos T/análise , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Humanos
15.
Mucosal Immunol ; 13(4): 626-636, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32066836

RESUMO

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.


Assuntos
Suscetibilidade a Doenças , Técnicas de Silenciamento de Genes , Imunoglobulinas/genética , Inflamação/etiologia , Inflamação/metabolismo , Receptores de Citocinas/genética , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Imunidade Adaptativa , Animais , Biomarcadores , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Marcação de Genes , Imunidade Inata , Imuno-Histoquímica , Inflamação/patologia , Camundongos , Camundongos Knockout , Mucosa Respiratória/patologia
16.
J Exp Med ; 216(12): 2689-2700, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31601676

RESUMO

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.


Assuntos
Fungos/fisiologia , Interações Microbianas , Micobioma , Receptores de Superfície Celular/metabolismo , Animais , Quitina/metabolismo , DNA Espaçador Ribossômico , Modelos Animais de Doenças , Enterite/etiologia , Enterite/metabolismo , Enterite/patologia , Microbioma Gastrointestinal , Expressão Gênica , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Metagenômica , Camundongos , Camundongos Transgênicos , Ligação Proteica , RNA Ribossômico 16S
17.
J Exp Med ; 216(12): 2714-2723, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31582416

RESUMO

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.


Assuntos
Imunidade Inata , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/metabolismo , Mucosa/imunologia , Mucosa/metabolismo , Muco/metabolismo , Animais , Proteção Cruzada/imunologia , Células Caliciformes/citologia , Células Caliciformes/metabolismo , Hiperplasia , Interleucina-13/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/parasitologia , Masculino , Camundongos , Camundongos Knockout , Mucinas/biossíntese , Trichinella spiralis/imunologia , Triquinelose/imunologia , Triquinelose/parasitologia
18.
Cell Rep ; 23(13): 3750-3758, 2018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29949760

RESUMO

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.


Assuntos
Tecido Linfoide/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Movimento Celular , Citrobacter rodentium/patogenicidade , Família 7 do Citocromo P450/metabolismo , Infecções por Enterobacteriaceae/patologia , Infecções por Enterobacteriaceae/prevenção & controle , Infecções por Enterobacteriaceae/veterinária , Humanos , Hidroxicolesteróis/química , Hidroxicolesteróis/metabolismo , Imunidade Inata , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Ligantes , Tecido Linfoide/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucosa/citologia , Mucosa/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/genética , Esteroide Hidroxilases/deficiência , Esteroide Hidroxilases/genética , Esteroide Hidroxilases/metabolismo
19.
PLoS One ; 13(11): e0207794, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30500852

RESUMO

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.


Assuntos
Vacinas contra a AIDS/imunologia , Antígenos CD40/imunologia , Células Dendríticas/imunologia , Epitopos de Linfócito T/imunologia , Anticorpos Anti-HIV/imunologia , HIV-1/imunologia , Receptores Imunológicos/imunologia , Animais , Macaca mulatta , Masculino , Terapia de Alvo Molecular
20.
Science ; 359(6379): 1056-1061, 2018 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-29496881

RESUMO

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.


Assuntos
Imunidade Adaptativa , Neurônios Adrenérgicos/imunologia , Imunidade Inata , Linfócitos/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Animais , Humanos , Inflamação/imunologia , Intestinos/imunologia , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Rede Nervosa/imunologia , Receptores Adrenérgicos beta 2/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa