Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 371
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 57(9): 2013-2029, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39151425

RESUMEN

The intestinal microbiota determines immune responses against extraintestinal antigens, including tumor-associated antigens. Indeed, depletion or gross perturbation of the microbiota undermines the efficacy of cancer immunotherapy, thereby compromising the clinical outcome of cancer patients. In this review, we discuss the long-distance effects of the gut microbiota and the mechanisms governing antitumor immunity, such as the translocation of intestinal microbes into tumors, migration of leukocyte populations from the gut to the rest of the body, including tumors, as well as immunomodulatory microbial products and metabolites. The relationship between these pathways is incompletely understood, in particular the significance of the tumor microbiota with respect to the identification of host and/or microbial products that regulate the egress of bacteria and immunocytes toward tumor beds.


Asunto(s)
Microbioma Gastrointestinal , Vigilancia Inmunológica , Neoplasias , Humanos , Neoplasias/inmunología , Neoplasias/microbiología , Neoplasias/terapia , Microbioma Gastrointestinal/inmunología , Animales , Vigilancia Inmunológica/inmunología , Inmunoterapia/métodos , Monitorización Inmunológica
2.
Nat Immunol ; 19(2): 173-182, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29311694

RESUMEN

CD8+ T cell immunosurveillance dynamics influence the outcome of intracellular infections and cancer. Here we used two-photon intravital microscopy to visualize the responses of CD8+ resident memory T cells (TRM cells) within the reproductive tracts of live female mice. We found that mucosal TRM cells were highly motile, but paused and underwent in situ division after local antigen challenge. TRM cell reactivation triggered the recruitment of recirculating memory T cells that underwent antigen-independent TRM cell differentiation in situ. However, the proliferation of pre-existing TRM cells dominated the local mucosal recall response and contributed most substantially to the boosted secondary TRM cell population. We observed similar results in skin. Thus, TRM cells can autonomously regulate the expansion of local immunosurveillance independently of central memory or proliferation in lymphoid tissue.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunidad Mucosa/inmunología , Memoria Inmunológica/inmunología , Vigilancia Inmunológica/inmunología , Membrana Mucosa/inmunología , Animales , Femenino , Microscopía Intravital , Ratones , Membrana Mucosa/citología , Piel/inmunología
3.
Nat Immunol ; 19(12): 1330-1340, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30420624

RESUMEN

Up to 49% of certain types of cancer are attributed to obesity, and potential mechanisms include overproduction of hormones, adipokines, and insulin. Cytotoxic immune cells, including natural killer (NK) cells and CD8+ T cells, are important in tumor surveillance, but little is known about the impact of obesity on immunosurveillance. Here, we show that obesity induces robust peroxisome proliferator-activated receptor (PPAR)-driven lipid accumulation in NK cells, causing complete 'paralysis' of their cellular metabolism and trafficking. Fatty acid administration, and PPARα and PPARδ (PPARα/δ) agonists, mimicked obesity and inhibited mechanistic target of rapamycin (mTOR)-mediated glycolysis. This prevented trafficking of the cytotoxic machinery to the NK cell-tumor synapse. Inhibiting PPARα/δ or blocking the transport of lipids into mitochondria reversed NK cell metabolic paralysis and restored cytotoxicity. In vivo, NK cells had blunted antitumor responses and failed to reduce tumor growth in obesity. Our results demonstrate that the lipotoxic obese environment impairs immunosurveillance and suggest that metabolic reprogramming of NK cells may improve cancer outcomes in obesity.


Asunto(s)
Vigilancia Inmunológica/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Melanoma Experimental/inmunología , Obesidad/inmunología , Adulto , Animales , Femenino , Humanos , Células Asesinas Naturales/patología , Masculino , Melanoma Experimental/complicaciones , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Obesidad/complicaciones , Adulto Joven
4.
Immunity ; 52(5): 872-884.e5, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32433950

RESUMEN

Some endocrine organs are frequent targets of autoimmune attack. Here, we addressed the origin of autoimmune disease from the viewpoint of feedback control. Endocrine tissues maintain mass through feedback loops that balance cell proliferation and removal according to hormone-driven regulatory signals. We hypothesized the existence of a dedicated mechanism that detects and removes mutant cells that missense the signal and therefore hyperproliferate and hypersecrete with potential to disrupt organismal homeostasis. In this mechanism, hypersecreting cells are preferentially eliminated by autoreactive T cells at the cost of a fragility to autoimmune disease. The "autoimmune surveillance of hypersecreting mutants" (ASHM) hypothesis predicts the presence of autoreactive T cells in healthy individuals and the nature of self-antigens as peptides from hormone secretion pathway. It explains why some tissues get prevalent autoimmune disease, whereas others do not and instead show prevalent mutant-expansion disease (e.g., hyperparathyroidism). The ASHM hypothesis is testable, and we discuss experimental follow-up.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Diabetes Mellitus Tipo 1/inmunología , Glándulas Endocrinas/inmunología , Sistema Endocrino/inmunología , Vigilancia Inmunológica/inmunología , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Proliferación Celular/genética , Proliferación Celular/fisiología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Glándulas Endocrinas/citología , Glándulas Endocrinas/metabolismo , Sistema Endocrino/citología , Sistema Endocrino/metabolismo , Femenino , Humanos , Vigilancia Inmunológica/genética , Masculino , Mutación , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
5.
Immunity ; 52(5): 794-807.e7, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32298648

RESUMEN

Lymphocyte homeostasis and immune surveillance require that T and B cells continuously recirculate between secondary lymphoid organs. Here, we used intravital microscopy to define lymphocyte trafficking routes within the spleen, an environment of open blood circulation and shear forces unlike other lymphoid organs. Upon release from arterioles into the red pulp sinuses, T cells latched onto perivascular stromal cells in a manner that was independent of the chemokine receptor CCR7 but sensitive to Gi protein-coupled receptor inhibitors. This latching sheltered T cells from blood flow and enabled unidirectional migration to the bridging channels and then to T zones, entry into which required CCR7. Inflammatory responses modified the chemotactic cues along the perivascular homing paths, leading to rapid block of entry. Our findings reveal a role for vascular structures in lymphocyte recirculation through the spleen, indicating the existence of separate entry and exit routes and that of a checkpoint located at the gate to the T zone.


Asunto(s)
Movimiento Celular/inmunología , Receptores CCR7/inmunología , Bazo/inmunología , Linfocitos T/inmunología , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Humanos , Vigilancia Inmunológica/inmunología , Microscopía Intravital , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Linfocitos/citología , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores CCR7/genética , Receptores CCR7/metabolismo , Transducción de Señal/inmunología , Bazo/citología , Bazo/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo
6.
Immunity ; 49(6): 1132-1147.e7, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30552022

RESUMEN

Serrated adenocarcinoma, an alternative pathway for colorectal cancer (CRC) development, accounts for 15%-30% of all CRCs and is aggressive and treatment resistant. We show that the expression of atypical protein kinase C ζ (PKCζ) and PKCλ/ι was reduced in human serrated tumors. Simultaneous inactivation of the encoding genes in the mouse intestinal epithelium resulted in spontaneous serrated tumorigenesis that progressed to advanced cancer with a strongly reactive and immunosuppressive stroma. Whereas epithelial PKCλ/ι deficiency led to immunogenic cell death and the infiltration of CD8+ T cells, which repressed tumor initiation, PKCζ loss impaired interferon and CD8+ T cell responses, which resulted in tumorigenesis. Combined treatment with a TGF-ß receptor inhibitor plus anti-PD-L1 checkpoint blockade showed synergistic curative activity. Analysis of human samples supported the relevance of these kinases in the immunosurveillance defects of human serrated CRC. These findings provide insight into avenues for the detection and treatment of this poor-prognosis subtype of CRC.


Asunto(s)
Mucosa Intestinal/inmunología , Neoplasias Intestinales/inmunología , Isoenzimas/inmunología , Proteína Quinasa C/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Femenino , Humanos , Vigilancia Inmunológica/genética , Vigilancia Inmunológica/inmunología , Mucosa Intestinal/enzimología , Mucosa Intestinal/patología , Neoplasias Intestinales/enzimología , Neoplasias Intestinales/genética , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo
7.
Immunity ; 46(1): 120-132, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28087238

RESUMEN

Lymphocytes circulate through lymph nodes (LN) in search for antigen in what is believed to be a continuous process. Here, we show that lymphocyte migration through lymph nodes and lymph occurred in a non-continuous, circadian manner. Lymphocyte homing to lymph nodes peaked at night onset, with cells leaving the tissue during the day. This resulted in strong oscillations in lymphocyte cellularity in lymph nodes and efferent lymphatic fluid. Using lineage-specific genetic ablation of circadian clock function, we demonstrated this to be dependent on rhythmic expression of promigratory factors on lymphocytes. Dendritic cell numbers peaked in phase with lymphocytes, with diurnal oscillations being present in disease severity after immunization to induce experimental autoimmune encephalomyelitis (EAE). These rhythms were abolished by genetic disruption of T cell clocks, demonstrating a circadian regulation of lymphocyte migration through lymph nodes with time-of-day of immunization being critical for adaptive immune responses weeks later.


Asunto(s)
Inmunidad Adaptativa/inmunología , Quimiotaxis de Leucocito/inmunología , Relojes Circadianos/inmunología , Vigilancia Inmunológica/inmunología , Linfocitos/inmunología , Traslado Adoptivo , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Immunity ; 46(1): 133-147, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28087239

RESUMEN

Immuno-surveillance networks operating at barrier sites are tuned by local tissue cues to ensure effective immunity. Site-specific commensal bacteria provide key signals ensuring host defense in the skin and gut. However, how the oral microbiome and tissue-specific signals balance immunity and regulation at the gingiva, a key oral barrier, remains minimally explored. In contrast to the skin and gut, we demonstrate that gingiva-resident T helper 17 (Th17) cells developed via a commensal colonization-independent mechanism. Accumulation of Th17 cells at the gingiva was driven in response to the physiological barrier damage that occurs during mastication. Physiological mechanical damage, via induction of interleukin 6 (IL-6) from epithelial cells, tailored effector T cell function, promoting increases in gingival Th17 cell numbers. These data highlight that diverse tissue-specific mechanisms govern education of Th17 cell responses and demonstrate that mechanical damage helps define the immune tone of this important oral barrier.


Asunto(s)
Encía/inmunología , Inmunidad Mucosa/inmunología , Vigilancia Inmunológica/inmunología , Mucosa Bucal/inmunología , Células Th17/inmunología , Animales , Citometría de Flujo , Encía/microbiología , Humanos , Masticación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota , Mucosa Bucal/microbiología , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Nature ; 577(7792): 689-694, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31942068

RESUMEN

Immune surveillance against pathogens and tumours in the central nervous system is thought to be limited owing to the lack of lymphatic drainage. However, the characterization of the meningeal lymphatic network has shed light on previously unappreciated ways that an immune response can be elicited to antigens that are expressed in the brain1-3. Despite progress in our understanding of the development and structure of the meningeal lymphatic system, the contribution of this network in evoking a protective antigen-specific immune response in the brain remains unclear. Here, using a mouse model of glioblastoma, we show that the meningeal lymphatic vasculature can be manipulated to mount better immune responses against brain tumours. The immunity that is mediated by CD8 T cells to the glioblastoma antigen is very limited when the tumour is confined to the central nervous system, resulting in uncontrolled tumour growth. However, ectopic expression of vascular endothelial growth factor C (VEGF-C) promotes enhanced priming of CD8 T cells in the draining deep cervical lymph nodes, migration of CD8 T cells into the tumour, rapid clearance of the glioblastoma and a long-lasting antitumour memory response. Furthermore, transfection of an mRNA construct that expresses VEGF-C works synergistically with checkpoint blockade therapy to eradicate existing glioblastoma. These results reveal the capacity of VEGF-C to promote immune surveillance of tumours, and suggest a new therapeutic approach to treat brain tumours.


Asunto(s)
Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Vigilancia Inmunológica/inmunología , Ganglios Linfáticos/inmunología , Vasos Linfáticos/inmunología , Factor C de Crecimiento Endotelial Vascular/metabolismo , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/inmunología , Línea Celular Tumoral , Movimiento Celular , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Reactividad Cruzada , Femenino , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Células HEK293 , Humanos , Memoria Inmunológica/inmunología , Linfangiogénesis , Masculino , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Meninges/inmunología , Ratones , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Factor C de Crecimiento Endotelial Vascular/administración & dosificación , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/uso terapéutico
10.
Immunity ; 45(6): 1270-1284, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-27939671

RESUMEN

Infections induce pathogen-specific T cell differentiation into diverse effectors (Teff) that give rise to memory (Tmem) subsets. The cell-fate decisions and lineage relationships that underlie these transitions are poorly understood. Here, we found that the chemokine receptor CX3CR1 identifies three distinct CD8+ Teff and Tmem subsets. Classical central (Tcm) and effector memory (Tem) cells and their corresponding Teff precursors were CX3CR1- and CX3CR1high, respectively. Viral infection also induced a numerically stable CX3CR1int subset that represented ∼15% of blood-borne Tmem cells. CX3CR1int Tmem cells underwent more frequent homeostatic divisions than other Tmem subsets and not only self-renewed, but also contributed to the expanding CX3CR1- Tcm pool. Both Tcm and CX3CR1int cells homed to lymph nodes, but CX3CR1int cells, and not Tem cells, predominantly surveyed peripheral tissues. As CX3CR1int Tmem cells present unique phenotypic, homeostatic, and migratory properties, we designate this subset peripheral memory (tpm) cells and propose that tpm cells are chiefly responsible for the global surveillance of non-lymphoid tissues.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Homeostasis/inmunología , Vigilancia Inmunológica/inmunología , Receptores de Quimiocina/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Receptor 1 de Quimiocinas CX3C , Separación Celular , Citometría de Flujo , Memoria Inmunológica/inmunología , Ratones , Ratones Endogámicos C57BL
11.
Immunity ; 44(4): 725-7, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27096315

RESUMEN

Non-alcoholic fatty liver disease is associated with hepatocellular carcinoma. In the March 10 issue of Nature, Greten and colleagues report that this metabolic disruption affects tumor surveillance by depleting CD4+ T helper cells through lipotoxic mechanisms associated with NAFLD.


Asunto(s)
Carcinoma Hepatocelular/inmunología , Supervivencia Celular/inmunología , Vigilancia Inmunológica/inmunología , Neoplasias Hepáticas/inmunología , Enfermedad del Hígado Graso no Alcohólico/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Humanos , Ratones , Enfermedad del Hígado Graso no Alcohólico/patología
12.
Nat Immunol ; 13(4): 343-51, 2012 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-22430787

RESUMEN

In the complex interplay between malignant cells and their microenvironment, caspase-1 activation complexes (inflammasomes) have contrasting roles. Inflammasomes may operate at the cell-autonomous level to eliminate malignant precursors through programmed cell death or, conversely, may stimulate the production of trophic factors for cancer cells and their stroma. In inflammatory cells, caspase-1 activation can fuel a cycle that leads to sterile inflammation and carcinogenesis, whereas in antigen-presenting cells, inflammasomes can stimulate anticancer immune responses. The inhibition of inflammasomes or neutralization of their products, mainly interleukin 1ß (IL-1ß) and IL-18, has profound effects on carcinogenesis and tumor progression. Thus, inflammasomes are promising therapeutic targets in cancer-related clinical conditions. Here we discuss present and future indications for the clinical use of inflammasome inhibitors.


Asunto(s)
Inflamasomas/inmunología , Neoplasias/inmunología , Animales , Humanos , Vigilancia Inmunológica/inmunología
13.
Nat Immunol ; 13(9): 832-42, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22842346

RESUMEN

The mechanisms by which tumor microenvironments modulate nucleic acid-mediated innate immunity remain unknown. Here we identify the receptor TIM-3 as key in circumventing the stimulatory effects of nucleic acids in tumor immunity. Tumor-associated dendritic cells (DCs) in mouse tumors and patients with cancer had high expression of TIM-3. DC-derived TIM-3 suppressed innate immune responses through the recognition of nucleic acids by Toll-like receptors and cytosolic sensors via a galectin-9-independent mechanism. In contrast, TIM-3 interacted with the alarmin HMGB1 to interfere with the recruitment of nucleic acids into DC endosomes and attenuated the therapeutic efficacy of DNA vaccination and chemotherapy by diminishing the immunogenicity of nucleic acids released from dying tumor cells. Our findings define a mechanism whereby tumor microenvironments suppress antitumor immunity mediated by nucleic acids.


Asunto(s)
Células Dendríticas/inmunología , Proteína HMGB1/inmunología , Inmunidad Innata , Neoplasias/inmunología , Ácidos Nucleicos/inmunología , Receptores Virales/inmunología , Microambiente Tumoral/inmunología , Animales , Células Dendríticas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Proteína HMGB1/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A , Humanos , Immunoblotting , Vigilancia Inmunológica/inmunología , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Microscopía Fluorescente , Neoplasias/metabolismo , Receptores de Reconocimiento de Patrones/inmunología , Receptores Virales/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Nat Immunol ; 13(9): 872-9, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22885985

RESUMEN

T cells bearing γδ T cell antigen receptors (TCRs) function in lymphoid stress surveillance. However, the contribution of γδ TCRs to such responses is unclear. Here we found that the TCR of a human V(γ)4V(δ)5 clone directly bound endothelial protein C receptor (EPCR), which allowed γδ T cells to recognize both endothelial cells targeted by cytomegalovirus and epithelial tumors. EPCR is a major histocompatibility complex-like molecule that binds lipids analogously to the antigen-presenting molecule CD1d. However, the V(γ)4V(δ)5 TCR bound EPCR independently of lipids, in an antibody-like way. Moreover, the recognition of target cells by γδ T cells required a multimolecular stress signature composed of EPCR and costimulatory ligand(s). Our results demonstrate how a γδ TCR mediates recognition of broadly stressed human cells by engaging a stress-regulated self antigen.


Asunto(s)
Antígenos CD/inmunología , Infecciones por Citomegalovirus/inmunología , Vigilancia Inmunológica/inmunología , Neoplasias Glandulares y Epiteliales/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores de Superficie Celular/inmunología , Estrés Fisiológico/inmunología , Antígenos CD/metabolismo , Citomegalovirus/inmunología , Receptor de Proteína C Endotelial , Humanos , Immunoblotting , Inmunoprecipitación , Unión Proteica , Receptores de Antígenos de Linfocitos T gamma-delta/química , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores de Superficie Celular/metabolismo , Subgrupos de Linfocitos T/química , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T/química , Linfocitos T/inmunología , Linfocitos T/metabolismo
15.
J Immunol ; 208(2): 227-234, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-35017212

RESUMEN

Our body's most outward facing epithelial barrier, the skin, serves as the frontline defense against myriad environmental assailants. To combat these motley threats, the skin has evolved a sophisticated immunological arsenal. In this article, I provide an overview of the skin's complex architecture and the distinct microniches in which immune cells reside and function. I review burgeoning literature on the synchronized immune, stromal, epithelial, and neuronal cell responses in healthy and inflamed skin. Next, I delve into the distinct requirement and mechanisms of long-term immune surveillance and tissue adaptation at the cutaneous frontier. Finally, by discussing the contributions of immune cells in maintaining and restoring tissue integrity, I underscore the constellation of noncanonical functions undertaken by the skin immune system. Just as our skin's immune system benefits from embracing diverse defense strategies, so, too, must we in the immunology research community support disparate perspectives and people from all walks of life.


Asunto(s)
Fenómenos del Sistema Inmunológico/fisiología , Vigilancia Inmunológica/inmunología , Piel/inmunología , Humanos , Sistema Inmunológico/inmunología , Piel/anatomía & histología , Uniones Estrechas/inmunología
16.
Immunity ; 41(2): 207-218, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25131531

RESUMEN

Coreceptor CD4 and CD8αß double-negative (DN) TCRαß(+) intraepithelial T cells, although numerous, have been greatly overlooked and their contribution to the immune response is not known. Here we used T cell receptor (TCR) sequencing of single cells combined with retrogenic expression of TCRs to study the fate and the major histocompatibility complex (MHC) restriction of DN TCRαß(+) intraepithelial T cells. The data show that commitment of thymic precursors to the DN TCRαß(+) lineage is imprinted by their TCR specificity. Moreover, the TCRs they express display a diverse and unusual pattern of MHC restriction that is nonoverlapping with that of CD4(+) or CD8αß(+) T cells, indicating that they sense antigens that are not recognized by the conventional T cell subsets. The new insights indicate that DN TCRαß(+) T cells form a third lineage of TCRαß T lymphocytes expressing a variable TCR repertoire, which serve nonredundant immune functions.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Linaje de la Célula/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Animales , Diferenciación Celular/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Vigilancia Inmunológica/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados
17.
Nat Immunol ; 11(9): 785-97, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20720586

RESUMEN

Nearly a century after the significance of the human complement system was recognized, we have come to realize that its functions extend far beyond the elimination of microbes. Complement acts as a rapid and efficient immune surveillance system that has distinct effects on healthy and altered host cells and foreign intruders. By eliminating cellular debris and infectious microbes, orchestrating immune responses and sending 'danger' signals, complement contributes substantially to homeostasis, but it can also take action against healthy cells if not properly controlled. This review describes our updated view of the function, structure and dynamics of the complement network, highlights its interconnection with immunity at large and with other endogenous pathways, and illustrates its multiple roles in homeostasis and disease.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Homeostasis/inmunología , Vigilancia Inmunológica/inmunología , Apoptosis/inmunología , Enfermedad , Regulación de la Expresión Génica , Humanos , Transducción de Señal
18.
J Immunol ; 204(2): 286-293, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31907271

RESUMEN

At steady state, the CNS parenchyma has few to no lymphocytes and less potent Ag-presentation capability compared with other organs. However, the meninges surrounding the CNS host diverse populations of immune cells that influence how CNS-related immune responses develop. Interstitial and cerebrospinal fluid produced in the CNS is continuously drained, and recent advances have emphasized that this process is largely taking place through the lymphatic system. To what extent this fluid process mobilizes CNS-derived Ags toward meningeal immune cells and subsequently the peripheral immune system through the lymphatic vessel network is a question of significant clinical importance for autoimmunity, tumor immunology, and infectious disease. Recent advances in understanding the role of meningeal lymphatics as a communicator between the brain and peripheral immunity are discussed in this review.


Asunto(s)
Encéfalo/inmunología , Vigilancia Inmunológica/inmunología , Vasos Linfáticos , Meninges/inmunología , Animales , Sistema Nervioso Central/inmunología , Humanos
19.
J Cell Mol Med ; 25(5): 2333-2341, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33605033

RESUMEN

Autophagy is frequently induced in the hypoxic tumour microenvironment. Accumulating evidence reveals important functions of autophagy at the tumour-immune interface. Herein, we propose an update on the roles of autophagy in modulating tumour immunity. Autophagy promotes adaptive resistance of established tumours to the cytotoxic effects of natural killer cells (NKs), macrophages and effector T cells. Increased autophagic flux in tumours dampen their immunogenicity and inhibits the expansion of cytotoxic T lymphocytes (CTLs) by suppressing the activation of STING type I interferon signalling (IFN-I) innate immune sensing pathway. Autophagy in suppressive tumour-infiltrating immune subsets maintains their survival through metabolic remodelling. On the other hand, autophagy is involved in the antigen processing and presentation process, which is essential for anti-tumour immune responses. Genetic deletion of autophagy induces spontaneous tumours in some models. Thus, the role of autophagy is context-dependent. In summary, our review has revealed the dichotomous roles of autophagy in modulating tumour immunity. Broad targeting of autophagy may not yield maximal benefits. The characterization of specific genes regulating tumour immunogenicity and innovation in targeted delivery of autophagy inhibitors into certain tumours are among the most urgent tasks to sensitize cold cancers to immunotherapy.


Asunto(s)
Autofagia , Inmunidad , Neoplasias/etiología , Neoplasias/metabolismo , Microambiente Tumoral/inmunología , Inmunidad Adaptativa , Animales , Presentación de Antígeno , Antígenos de Neoplasias , Autofagia/genética , Autofagia/inmunología , Biomarcadores , Susceptibilidad a Enfermedades , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunidad Innata , Vigilancia Inmunológica/genética , Vigilancia Inmunológica/inmunología , Neoplasias/patología , Transducción de Señal , Microambiente Tumoral/genética
20.
Eur J Immunol ; 50(3): 439-444, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31729760

RESUMEN

It is well established that therapeutic impairment of Foxp3+ Treg in mice and humans favors immune rejection of solid tumors. Less explored is the impact Foxp3 allelic variants may have on tumor incidence, progression and therapy. In this work, we tested and demonstrate that the Foxp3fgfp reporter allele, found previously to either enhance or reduce Treg function in specific autoimmunity settings, confers increased anti-tumor immunity. Our conclusions stem out of the analysis of three tumor models of different tissue origin, in two murine genetic backgrounds. When compared to wild type animals, mice carrying the Foxp3fgfp allele spontaneously delay, reduce or prevent primary tumor growth, decrease metastasis growth, and potentiate the response to anti-CTLA4 monotherapy. These findings suggest allelic variances at the Foxp3 locus may serve as predictive indicators for personalized therapy and prognostics, and point at possible new therapeutic targets.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Vigilancia Inmunológica/genética , Neoplasias Experimentales/inmunología , Alelos , Animales , Factores de Transcripción Forkhead/genética , Vigilancia Inmunológica/inmunología , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA