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










Intervalo de año de publicación
1.
Science ; 384(6692): eadk6200, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38574174

RESUMEN

Males and females exhibit profound differences in immune responses and disease susceptibility. However, the factors responsible for sex differences in tissue immunity remain poorly understood. Here, we uncovered a dominant role for type 2 innate lymphoid cells (ILC2s) in shaping sexual immune dimorphism within the skin. Mechanistically, negative regulation of ILC2s by androgens leads to a reduction in dendritic cell accumulation and activation in males, along with reduced tissue immunity. Collectively, our results reveal a role for the androgen-ILC2-dendritic cell axis in controlling sexual immune dimorphism. Moreover, this work proposes that tissue immune set points are defined by the dual action of sex hormones and the microbiota, with sex hormones controlling the strength of local immunity and microbiota calibrating its tone.


Asunto(s)
Andrógenos , Células Dendríticas , Inmunidad Innata , Linfocitos , Caracteres Sexuales , Piel , Femenino , Masculino , Andrógenos/metabolismo , Células Dendríticas/inmunología , Hormonas Esteroides Gonadales/metabolismo , Linfocitos/inmunología , Piel/inmunología , Animales , Ratones , Ratones Endogámicos C57BL , Microbiota
2.
Proc Natl Acad Sci U S A ; 121(11): e2322574121, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38451947

RESUMEN

The somatosensory nervous system surveils external stimuli at barrier tissues, regulating innate immune cells under infection and inflammation. The roles of sensory neurons in controlling the adaptive immune system, and more specifically immunity to the microbiota, however, remain elusive. Here, we identified a mechanism for direct neuroimmune communication between commensal-specific T lymphocytes and somatosensory neurons mediated by the neuropeptide calcitonin gene-related peptide (CGRP) in the skin. Intravital imaging revealed that commensal-specific T cells are in close proximity to cutaneous nerve fibers in vivo. Correspondingly, we observed upregulation of the receptor for the neuropeptide CGRP, RAMP1, in CD8+ T lymphocytes induced by skin commensal colonization. The neuroimmune CGRP-RAMP1 signaling axis functions in commensal-specific T cells to constrain Type 17 responses and moderate the activation status of microbiota-reactive lymphocytes at homeostasis. As such, modulation of neuroimmune CGRP-RAMP1 signaling in commensal-specific T cells shapes the overall activation status of the skin epithelium, thereby impacting the outcome of responses to insults such as wounding. The ability of somatosensory neurons to control adaptive immunity to the microbiota via the CGRP-RAMP1 axis underscores the various layers of regulation and multisystem coordination required for optimal microbiota-reactive T cell functions under steady state and pathology.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina , Neuroinmunomodulación , Péptido Relacionado con Gen de Calcitonina/genética , Proteína 1 Modificadora de la Actividad de Receptores/genética , Receptores de Péptido Relacionado con el Gen de Calcitonina , Inmunidad Adaptativa
3.
Proc Natl Acad Sci U S A ; 120(49): e2304905120, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38011570

RESUMEN

Mild or transient dietary restriction (DR) improves many aspects of health and aging. Emerging evidence from us and others has demonstrated that DR also optimizes the development and quality of immune responses. However, the factors and mechanisms involved remain to be elucidated. Here, we propose that DR-induced optimization of immunological memory requires a complex cascade of events involving memory T cells, the intestinal microbiota, and myeloid cells. Our findings suggest that DR enhances the ability of memory T cells to recruit and activate myeloid cells in the context of a secondary infection. Concomitantly, DR promotes the expansion of commensal Bifidobacteria within the large intestine, which produce the short-chain fatty acid acetate. Acetate conditioning of the myeloid compartment during DR enhances the capacity of these cells to kill pathogens. Enhanced host protection during DR is compromised when Bifidobacteria expansion is prevented, indicating that microbiota configuration and function play an important role in determining immune responsiveness to this dietary intervention. Altogether, our study supports the idea that DR induces both memory T cells and the gut microbiota to produce distinct factors that converge on myeloid cells to promote optimal pathogen control. These findings suggest that nutritional cues can promote adaptation and co-operation between multiple immune cells and the gut microbiota, which synergize to optimize immunity and protect the collective metaorganism.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Ácidos Grasos Volátiles , Acetatos
4.
Cell ; 186(3): 607-620.e17, 2023 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-36640762

RESUMEN

Tissue immunity and responses to injury depend on the coordinated action and communication among physiological systems. Here, we show that, upon injury, adaptive responses to the microbiota directly promote sensory neuron regeneration. At homeostasis, tissue-resident commensal-specific T cells colocalize with sensory nerve fibers within the dermis, express a transcriptional program associated with neuronal interaction and repair, and promote axon growth and local nerve regeneration following injury. Mechanistically, our data reveal that the cytokine interleukin-17A (IL-17A) released by commensal-specific Th17 cells upon injury directly signals to sensory neurons via IL-17 receptor A, the transcription of which is specifically upregulated in injured neurons. Collectively, our work reveals that in the context of tissue damage, preemptive immunity to the microbiota can rapidly bridge biological systems by directly promoting neuronal repair, while also identifying IL-17A as a major determinant of this fundamental process.


Asunto(s)
Interleucina-17 , Microbiota , Regeneración Nerviosa , Células Th17 , Axones , Regeneración Nerviosa/fisiología , Células Receptoras Sensoriales , Animales , Ratones , Células Th17/citología
5.
bioRxiv ; 2023 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-38234748

RESUMEN

The somatosensory nervous system surveils external stimuli at barrier tissues, regulating innate immune cells under infection and inflammation. The roles of sensory neurons in controlling the adaptive immune system, and more specifically immunity to the microbiota, however, remain elusive. Here, we identified a novel mechanism for direct neuroimmune communication between commensal-specific T lymphocytes and somatosensory neurons mediated by the neuropeptide Calcitonin Gene-Related Peptide (CGRP) in the skin. Intravital imaging revealed that commensal-specific T cells are in close proximity to cutaneous nerve fibers in vivo . Correspondingly, we observed upregulation of the receptor for the neuropeptide CGRP, RAMP1, in CD8 + T lymphocytes induced by skin commensal colonization. Neuroimmune CGRP-RAMP1 signaling axis functions in commensal-specific T cells to constrain Type 17 responses and moderate the activation status of microbiota-reactive lymphocytes at homeostasis. As such, modulation of neuroimmune CGRP-RAMP1 signaling in commensal-specific T cells shapes the overall activation status of the skin epithelium, thereby impacting the outcome of responses to insults such as wounding. The ability of somatosensory neurons to control adaptive immunity to the microbiota via the CGRP-RAMP1 axis underscores the various layers of regulation and multisystem coordination required for optimal microbiota-reactive T cell functions under steady state and pathology. Significance statement: Multisystem coordination at barrier surfaces is critical for optimal tissue functions and integrity, in response to microbial and environmental cues. In this study, we identified a novel neuroimmune crosstalk mechanism between the sensory nervous system and the adaptive immune response to the microbiota, mediated by the neuropeptide CGRP and its receptor RAMP1 on skin microbiota-induced T lymphocytes. The neuroimmune CGPR-RAMP1 axis constrains adaptive immunity to the microbiota and overall limits the activation status of the skin epithelium, impacting tissue responses to wounding. Our study opens the door to a new avenue to modulate adaptive immunity to the microbiota utilizing neuromodulators, allowing for a more integrative and tailored approach to harnessing microbiota-induced T cells to promote barrier tissue protection and repair.

6.
Cell Rep ; 38(1): 110184, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34986349

RESUMEN

MV130 is an inactivated polybacterial mucosal vaccine that confers protection to patients against recurrent respiratory infections, including those of viral etiology. However, its mechanism of action remains poorly understood. Here, we find that intranasal prophylaxis with MV130 modulates the lung immune landscape and provides long-term heterologous protection against viral respiratory infections in mice. Intranasal administration of MV130 provides protection against systemic candidiasis in wild-type and Rag1-deficient mice lacking functional lymphocytes, indicative of innate immune-mediated protection. Moreover, pharmacological inhibition of trained immunity with metformin abrogates the protection conferred by MV130 against influenza A virus respiratory infection. MV130 induces reprogramming of both mouse bone marrow progenitor cells and in vitro human monocytes, promoting an enhanced cytokine production that relies on a metabolic shift. Our results unveil that the mucosal administration of a fully inactivated bacterial vaccine provides protection against viral infections by a mechanism associated with the induction of trained immunity.


Asunto(s)
Vacunas Bacterianas/inmunología , Inmunidad Mucosa/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Mucosa Respiratoria/inmunología , Infecciones del Sistema Respiratorio/prevención & control , Administración Intranasal , Animales , Anticuerpos Antivirales/inmunología , Bacterias/inmunología , Vacunas Bacterianas/administración & dosificación , Candidiasis/prevención & control , Línea Celular , Chlorocebus aethiops , Citocinas/biosíntesis , Humanos , Virus de la Influenza A/inmunología , Células L , Pulmón/inmunología , Metformina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/virología , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología
7.
Cell ; 184(14): 3794-3811.e19, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34166614

RESUMEN

The microbiota plays a fundamental role in regulating host immunity. However, the processes involved in the initiation and regulation of immunity to the microbiota remain largely unknown. Here, we show that the skin microbiota promotes the discrete expression of defined endogenous retroviruses (ERVs). Keratinocyte-intrinsic responses to ERVs depended on cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes protein (STING) signaling and promoted the induction of commensal-specific T cells. Inhibition of ERV reverse transcription significantly impacted these responses, resulting in impaired immunity to the microbiota and its associated tissue repair function. Conversely, a lipid-enriched diet primed the skin for heightened ERV- expression in response to commensal colonization, leading to increased immune responses and tissue inflammation. Together, our results support the idea that the host may have co-opted its endogenous virome as a means to communicate with the exogenous microbiota, resulting in a multi-kingdom dialog that controls both tissue homeostasis and inflammation.


Asunto(s)
Retrovirus Endógenos/fisiología , Homeostasis , Inflamación/microbiología , Inflamación/patología , Microbiota , Animales , Bacterias/metabolismo , Cromosomas Bacterianos/genética , Dieta Alta en Grasa , Inflamación/inmunología , Inflamación/virología , Interferón Tipo I/metabolismo , Queratinocitos/metabolismo , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Nucleotidiltransferasas/metabolismo , Retroelementos/genética , Transducción de Señal , Piel/inmunología , Piel/microbiología , Linfocitos T/inmunología , Transcripción Genética
9.
J Immunother Cancer ; 9(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33980589

RESUMEN

BACKGROUND: Conventional type 1 dendritic cells (cDC1s) are central to antitumor immunity and their presence in the tumor microenvironment associates with improved outcomes in patients with cancer. DNGR-1 (CLEC9A) is a dead cell-sensing receptor highly restricted to cDC1s. DNGR-1 has been involved in both cross-presentation of dead cell-associated antigens and processes of disease tolerance, but its role in antitumor immunity has not been clarified yet. METHODS: B16 and MC38 tumor cell lines were inoculated subcutaneously into wild-type (WT) and DNGR-1-deficient mice. To overexpress Flt3L systemically, we performed gene therapy through the hydrodynamic injection of an Flt3L-encoding plasmid. To characterize the immune response, we performed flow cytometry and RNA-Seq of tumor-infiltrating cDC1s. RESULTS: Here, we found that cross-presentation of tumor antigens in the steady state was DNGR-1-independent. However, on Flt3L systemic overexpression, tumor growth was delayed in DNGR-1-deficient mice compared with WT mice. Of note, this protection was recapitulated by anti-DNGR-1-blocking antibodies in mice following Flt3L gene therapy. This improved antitumor immunity was associated with Batf3-dependent enhanced accumulation of CD8+ T cells and cDC1s within tumors. Mechanistically, the deficiency in DNGR-1 boosted an Flt3L-induced specific inflammatory gene signature in cDC1s, including Ccl5 expression. Indeed, the increased infiltration of cDC1s within tumors and their protective effect rely on CCL5/CCR5 chemoattraction. Moreover, FLT3LG and CCL5 or CCR5 gene expression signatures correlate with an enhanced cDC1 signature and a favorable overall survival in patients with cancer. Notably, cyclophosphamide elevated serum Flt3L levels and, in combination with the absence of DNGR-1, synergized against tumor growth. CONCLUSION: DNGR-1 limits the accumulation of tumor-infiltrating cDC1s promoted by Flt3L. Thus, DNGR-1 blockade may improve antitumor immunity in tumor therapy settings associated to high Flt3L expression.


Asunto(s)
Neoplasias del Colon/terapia , Células Dendríticas/metabolismo , Terapia Genética , Lectinas Tipo C/metabolismo , Melanoma Experimental/terapia , Proteínas de la Membrana/metabolismo , Receptores Inmunológicos/metabolismo , Neoplasias Cutáneas/terapia , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Técnicas de Cocultivo , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Neoplasias del Colon/metabolismo , Células Dendríticas/inmunología , Regulación Neoplásica de la Expresión Génica , Lectinas Tipo C/genética , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Receptores CCR5/genética , Receptores CCR5/metabolismo , Receptores Inmunológicos/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Carga Tumoral , Escape del Tumor , Microambiente Tumoral
10.
Cell ; 184(3): 615-627.e17, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33453153

RESUMEN

The microbiota shields the host against infections in a process known as colonization resistance. How infections themselves shape this fundamental process remains largely unknown. Here, we show that gut microbiota from previously infected hosts display enhanced resistance to infection. This long-term functional remodeling is associated with altered bile acid metabolism leading to the expansion of taxa that utilize the sulfonic acid taurine. Notably, supplying exogenous taurine alone is sufficient to induce this alteration in microbiota function and enhance resistance. Mechanistically, taurine potentiates the microbiota's production of sulfide, an inhibitor of cellular respiration, which is key to host invasion by numerous pathogens. As such, pharmaceutical sequestration of sulfide perturbs the microbiota's composition and promotes pathogen invasion. Together, this work reveals a process by which the host, triggered by infection, can deploy taurine as a nutrient to nourish and train the microbiota, promoting its resistance to subsequent infection.


Asunto(s)
Microbioma Gastrointestinal , Interacciones Huésped-Patógeno , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Recuento de Colonia Microbiana , Microbioma Gastrointestinal/efectos de los fármacos , Interacciones Huésped-Patógeno/efectos de los fármacos , Inmunidad , Ratones Endogámicos C57BL , Sulfuros/metabolismo , Taurina/farmacología
11.
Nat Immunol ; 20(10): 1311-1321, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31527833

RESUMEN

Whether screening the metabolic activity of immune cells facilitates discovery of molecular pathology remains unknown. Here we prospectively screened the extracellular acidification rate as a measure of glycolysis and the oxygen consumption rate as a measure of mitochondrial respiration in B cells from patients with primary antibody deficiency. The highest oxygen consumption rate values were detected in three study participants with persistent polyclonal B cell lymphocytosis (PPBL). Exome sequencing identified germline mutations in SDHA, which encodes succinate dehydrogenase subunit A, in all three patients with PPBL. SDHA gain-of-function led to an accumulation of fumarate in PPBL B cells, which engaged the KEAP1-Nrf2 system to drive the transcription of genes encoding inflammatory cytokines. In a single patient trial, blocking the activity of the cytokine interleukin-6 in vivo prevented systemic inflammation and ameliorated clinical disease. Overall, our study has identified pathological mitochondrial retrograde signaling as a disease modifier in primary antibody deficiency.


Asunto(s)
Linfocitos B/inmunología , Complejo II de Transporte de Electrones/genética , Inflamación/metabolismo , Linfocitosis/inmunología , Mitocondrias/metabolismo , Mutación/genética , Antiinflamatorios/farmacología , Respiración de la Célula , Células Cultivadas , Fumaratos/metabolismo , Glucólisis , Humanos , Inflamación/genética , Interleucina-6/antagonistas & inhibidores , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Consumo de Oxígeno , Estudios Prospectivos , Transducción de Señal , Secuenciación del Exoma
12.
Cell ; 178(5): 1088-1101.e15, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31442402

RESUMEN

Mammals evolved in the face of fluctuating food availability. How the immune system adapts to transient nutritional stress remains poorly understood. Here, we show that memory T cells collapsed in secondary lymphoid organs in the context of dietary restriction (DR) but dramatically accumulated within the bone marrow (BM), where they adopted a state associated with energy conservation. This response was coordinated by glucocorticoids and associated with a profound remodeling of the BM compartment, which included an increase in T cell homing factors, erythropoiesis, and adipogenesis. Adipocytes, as well as CXCR4-CXCL12 and S1P-S1P1R interactions, contributed to enhanced T cell accumulation in BM during DR. Memory T cell homing to BM during DR was associated with enhanced protection against infections and tumors. Together, this work uncovers a fundamental host strategy to sustain and optimize immunological memory during nutritional challenges that involved a temporal and spatial reorganization of the memory pool within "safe haven" compartments.


Asunto(s)
Médula Ósea/metabolismo , Memoria Inmunológica , Animales , Médula Ósea/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Restricción Calórica/veterinaria , Línea Celular Tumoral , Quimiocina CXCL12/metabolismo , Dieta Reductora/veterinaria , Metabolismo Energético , Regulación de la Expresión Génica , Glucocorticoides , Melanoma Experimental/mortalidad , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores CXCR4/metabolismo , Tasa de Supervivencia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
13.
Science ; 363(6422)2019 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-30523076

RESUMEN

Barrier tissues are primary targets of environmental stressors and are home to the largest number of antigen-experienced lymphocytes in the body, including commensal-specific T cells. We found that skin-resident commensal-specific T cells harbor a paradoxical program characterized by a type 17 program associated with a poised type 2 state. Thus, in the context of injury and exposure to inflammatory mediators such as interleukin-18, these cells rapidly release type 2 cytokines, thereby acquiring contextual functions. Such acquisition of a type 2 effector program promotes tissue repair. Aberrant type 2 responses can also be unleashed in the context of local defects in immunoregulation. Thus, commensal-specific T cells co-opt tissue residency and cell-intrinsic flexibility as a means to promote both local immunity and tissue adaptation to injury.


Asunto(s)
Plasticidad de la Célula , Piel/lesiones , Piel/microbiología , Simbiosis , Células Th17/inmunología , Células Th17/microbiología , Heridas y Lesiones/inmunología , Alarminas/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/microbiología , Candida albicans , Femenino , Factor de Transcripción GATA3/metabolismo , Interleucinas/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Microscopía Fluorescente , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Análisis de Secuencia de ARN , Staphylococcus epidermidis , Transcriptoma
14.
Science ; 362(6412): 351-356, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30337411

RESUMEN

Host injury triggers feedback mechanisms that limit tissue damage. Conventional type 1 dendritic cells (cDC1s) express dendritic cell natural killer lectin group receptor-1 (DNGR-1), encoded by the gene Clec9a, which senses tissue damage and favors cross-presentation of dead-cell material to CD8+ T cells. Here we find that DNGR-1 additionally reduces host-damaging inflammatory responses induced by sterile and infectious tissue injury in mice. DNGR-1 deficiency leads to exacerbated caerulein-induced necrotizing pancreatitis and increased pathology during systemic Candida albicans infection without affecting fungal burden. This effect is B and T cell-independent and attributable to increased neutrophilia in DNGR-1-deficient settings. Mechanistically, DNGR-1 engagement activates SHP-1 and inhibits MIP-2 (encoded by Cxcl2) production by cDC1s during Candida infection. This consequently restrains neutrophil recruitment and promotes disease tolerance. Thus, DNGR-1-mediated sensing of injury by cDC1s serves as a rheostat for the control of tissue damage, innate immunity, and immunopathology.


Asunto(s)
Candida albicans/inmunología , Candidiasis/patología , Células Dendríticas/inmunología , Lectinas Tipo C/fisiología , Infiltración Neutrófila/inmunología , Páncreas/patología , Pancreatitis Aguda Necrotizante/patología , Receptores Inmunológicos/fisiología , Animales , Lectinas Tipo C/genética , Ratones , Ratones Mutantes , Necrosis , Infiltración Neutrófila/genética , Páncreas/inmunología , Páncreas/microbiología , Pancreatitis Aguda Necrotizante/microbiología , Receptores Inmunológicos/genética
15.
Front Immunol ; 9: 1751, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30108585

RESUMEN

Tissue-resident memory CD8+ T (Trm) cells define a distinct non-recirculating subset. Trm cells constitute a first line of defense against local infections in barrier tissues, but they are also found in non-barrier tissues and play a role in antitumor immunity. Their differentiation in tissues and their phenotypical, transcriptional, and functional characteristics are the object of active research. Herein, we will discuss the potential existence of committed CD8+ Trm precursors and the genealogy of memory CD8+ T cell subsets. In addition to the priming of naive T cells, there is some plasticity of antigen-experienced effector and memory T cell subsets to generate Trm precursors. Local inflammation, antigen presentation, and cytokines drive Trm differentiation. It is of prime interest how specific dendritic cell subsets modulate priming and differentiation of Trm cells, as well as their reactivation within tissues. Research on how we can manipulate generation of memory T cells subsets is key for improved vaccination strategies.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Memoria Inmunológica/inmunología , Subgrupos de Linfocitos T/inmunología , Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/citología , Linaje de la Célula/inmunología , Plasticidad de la Célula/inmunología , Células Dendríticas/citología , Humanos , Modelos Inmunológicos , Subgrupos de Linfocitos T/citología
16.
Cell Rep ; 24(7): 1738-1746, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30110631

RESUMEN

The rapid transit from hypoxia to normoxia in the lung that follows the first breath in newborn mice coincides with alveolar macrophage (AM) differentiation. However, whether sensing of oxygen affects AM maturation and function has not been previously explored. We have generated mice whose AMs show a deficient ability to sense oxygen after birth by deleting Vhl, a negative regulator of HIF transcription factors, in the CD11c compartment (CD11cΔVhl mice). VHL-deficient AMs show an immature-like phenotype and an impaired self-renewal capacity in vivo that persists upon culture ex vivo. VHL-deficient phenotype is intrinsic in AMs derived from monocyte precursors in mixed bone marrow chimeras. Moreover, unlike control Vhlfl/fl, AMs from CD11cΔVhl mice do not reverse pulmonary alveolar proteinosis when transplanted into Csf2rb-/- mice, demonstrating that VHL contributes to AM-mediated surfactant clearance. Thus, our results suggest that optimal AM terminal differentiation, self-renewal, and homeostatic function requires their intact oxygen-sensing capacity.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular/genética , Hipoxia/genética , Macrófagos Alveolares/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Animales , Antígenos de Diferenciación Mielomonocítica/genética , Antígenos de Diferenciación Mielomonocítica/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Antígenos CD11/genética , Antígenos CD11/metabolismo , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Subunidad beta Común de los Receptores de Citocinas/deficiencia , Subunidad beta Común de los Receptores de Citocinas/genética , Eliminación de Gen , Regulación de la Expresión Génica , Humanos , Hipoxia/metabolismo , Hipoxia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Pulmón/metabolismo , Pulmón/patología , Macrófagos Alveolares/patología , Macrófagos Alveolares/trasplante , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxígeno/farmacología , Receptores de IgG/genética , Receptores de IgG/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico , Transducción de Señal , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
17.
Cancer Immunol Res ; 6(7): 798-811, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29678874

RESUMEN

T and NK lymphocytes express CD137 (4-1BB), a costimulatory receptor of the TNFR family whose function is exploitable for cancer immunotherapy. Mitochondria regulate the function and survival of T lymphocytes. Herein, we show that CD137 costimulation provided by agonist mAb and CD137L (4-1BBL) induced mitochondria enlargement that resulted in enhanced mitochondrial mass and transmembrane potential in human and mouse CD8+ T cells. Such mitochondrial changes increased T-cell respiratory capacities and were critically dependent on mitochondrial fusion protein OPA-1 expression. Mass and function of mitochondria in tumor-reactive CD8+ T cells from cancer-bearing mice were invigorated by agonist mAb to CD137, whereas mitochondrial baseline mass and function were depressed in CD137-deficient tumor reactive T cells. Tumor rejection induced by the synergistic combination of adoptive T-cell therapy and agonistic anti-CD137 was critically dependent on OPA-1 expression in transferred CD8+ T cells. Moreover, stimulation of CD137 with CD137 mAb in short-term cultures of human tumor-infiltrating lymphocytes led to mitochondria enlargement and increased transmembrane potential. Collectively, these data point to a critical link between mitochondrial morphology and function and enhanced antitumor effector activity upon CD137 costimulation of T cells. Cancer Immunol Res; 6(7); 798-811. ©2018 AACR.


Asunto(s)
Ligando 4-1BB/metabolismo , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Linfocitos T/metabolismo , Ligando 4-1BB/genética , Animales , Biomarcadores , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Citocinas/metabolismo , Citotoxicidad Inmunológica , Femenino , Silenciador del Gen , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental , Potencial de la Membrana Mitocondrial , Ratones , Ratones Noqueados , Mitocondrias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , ARN Interferente Pequeño/genética , Linfocitos T/inmunología , Microambiente Tumoral/inmunología
18.
J Immunol ; 200(10): 3475-3484, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29618524

RESUMEN

IL-2 is critical for peripheral tolerance mediated by regulatory T (Treg) cells, which represent an obstacle for effective cancer immunotherapy. Although IL-2 is important for effector (E) T cell function, it has been hypothesized that therapies blocking IL-2 signals weaken Treg cell activity, promoting immune responses. This hypothesis has been partially tested using anti-IL-2 or anti-IL-2R Abs with antitumor effects that cannot be exclusively attributed to lack of IL-2 signaling in vivo. In this work, we pursued an alternative strategy to block IL-2 signaling in vivo, taking advantage of the trimeric structure of the IL-2R. We designed an IL-2 mutant that conserves the capacity to bind to the αß-chains of the IL-2R but not to the γc-chain, thus having a reduced signaling capacity. We show our IL-2 mutein inhibits IL-2 Treg cell-dependent differentiation and expansion. Moreover, treatment with IL-2 mutein reduces Treg cell numbers and impairs tumor growth in mice. A mathematical model was used to better understand the effect of the mutein on Treg and E T cells, suggesting suitable strategies to improve its design. Our results show that it is enough to transiently inhibit IL-2 signaling to bias E and Treg cell balance in vivo toward immunity.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Interleucina-2/antagonistas & inhibidores , Linfocinas/farmacología , Neoplasias/terapia , Transducción de Señal/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Femenino , Humanos , Inmunoterapia/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/metabolismo , Tolerancia Periférica/efectos de los fármacos
19.
Nat Commun ; 8: 16073, 2017 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-28714465

RESUMEN

The goal of successful anti-tumoural immunity is the development of long-term protective immunity to prevent relapse. Infiltration of tumours with CD8+ T cells with a resident memory (Trm) phenotype correlates with improved survival. However, the interplay of circulating CD8+ T cells and Trm cells remains poorly explored in tumour immunity. Using different vaccination strategies that fine-tune the generation of Trm cells or circulating memory T cells, here we show that, while both subsets are sufficient for anti-tumour immunity, the presence of Trm cells improves anti-tumour efficacy. Transferred central memory T cells (Tcm) generate Trm cells following viral infection or tumour challenge. Anti-PD-1 treatment promotes infiltration of transferred Tcm cells within tumours, improving anti-tumour immunity. Moreover, Batf3-dependent dendritic cells are essential for reactivation of circulating memory anti-tumour response. Our findings show the plasticity, collaboration and requirements for reactivation of memory CD8+ T cells subsets needed for optimal tumour vaccination and immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/inmunología , Subgrupos de Linfocitos T/inmunología , Vaccinia/inmunología , Adaptación Fisiológica/inmunología , Animales , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Linfocitos T CD8-positivos/efectos de los fármacos , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Ratones , Ratones Noqueados , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Proteínas Represoras/genética , Subgrupos de Linfocitos T/efectos de los fármacos , Virus Vaccinia
20.
Front Immunol ; 8: 527, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28533780

RESUMEN

Sensing of microbe-associated molecular patterns or danger signals by innate immune receptors drives a complex exchange of information. Innate receptor signaling not only triggers transcriptional events but also induces profound changes in metabolic fluxes, redox balance, and metabolite abundance thereby influencing immune cell function. Mitochondria are at the core of metabolic adaptation to the changing environment. The close interaction between mitochondrial metabolism and immune signaling has emerged as a central regulator of innate sensing. Metabolic processes generate a constant flow of electrons that eventually end up in the mitochondrial electron transport chain (ETC). Two electron carriers and four respiratory complexes that can assemble as larger molecular supercomplexes compose the ETC in the mitochondrial inner membrane. While the meaning and biological relevance of such structural organization is a matter of passionate debates, recent data support that innate stimuli remodel the ETC. We will review the function of mitochondrial metabolism and ETC dynamics as innate rheostats that regulate signaling, transcription, and epigenetics to orchestrate innate immune responses.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...