Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunity ; 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38697116

RESUMEN

Cytokine expression during T cell differentiation is a highly regulated process that involves long-range promoter-enhancer and CTCF-CTCF contacts at cytokine loci. Here, we investigated the impact of dynamic chromatin loop formation within the topologically associating domain (TAD) in regulating the expression of interferon gamma (IFN-γ) and interleukin-22 (IL-22); these cytokine loci are closely located in the genome and are associated with complex enhancer landscapes, which are selectively active in type 1 and type 3 lymphocytes. In situ Hi-C analyses revealed inducible TADs that insulated Ifng and Il22 enhancers during Th1 cell differentiation. Targeted deletion of a 17 bp boundary motif of these TADs imbalanced Th1- and Th17-associated immunity, both in vitro and in vivo, upon Toxoplasma gondii infection. In contrast, this boundary element was dispensable for cytokine regulation in natural killer cells. Our findings suggest that precise cytokine regulation relies on lineage- and developmental stage-specific interactions of 3D chromatin architectures and enhancer landscapes.

2.
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
3.
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
4.
Immunol Rev ; 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38445769

RESUMEN

Maternal environmental exposures, particularly during gestation and lactation, significantly influence the immunological development and long-term immunity of offspring. Mammalian immune systems develop through crucial inputs from the environment, beginning in utero and continuing after birth. These critical developmental windows are essential for proper immune system development and, once closed, may not be reopened. This review focuses on the mechanisms by which maternal exposures, particularly to pathogens, diet, and microbiota, impact offspring immunity. Mechanisms driving maternal-offspring immune crosstalk include transfer of maternal antibodies, changes in the maternal microbiome and microbiota-derived metabolites, and transfer of immune cells and cytokines via the placenta and breastfeeding. We further discuss the role of transient maternal infections, which are common during pregnancy, in providing tissue-specific immune education to offspring. We propose a "maternal-driven immune education" hypothesis, which suggests that offspring can use maternal encounters that occur during a critical developmental window to develop optimal immune fitness against infection and inflammation.

5.
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
6.
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.

7.
J Clin Invest ; 131(17)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34623332

RESUMEN

We studied a child with severe viral, bacterial, fungal, and parasitic diseases, who was homozygous for a loss-of-function mutation of REL, encoding c-Rel, which is selectively expressed in lymphoid and myeloid cells. The patient had low frequencies of NK, effector memory cells reexpressing CD45RA (Temra) CD8+ T cells, memory CD4+ T cells, including Th1 and Th1*, Tregs, and memory B cells, whereas the counts and proportions of other leukocyte subsets were normal. Functional deficits of myeloid cells included the abolition of IL-12 and IL-23 production by conventional DC1s (cDC1s) and monocytes, but not cDC2s. c-Rel was also required for induction of CD86 expression on, and thus antigen-presenting cell function of, cDCs. Functional deficits of lymphoid cells included reduced IL-2 production by naive T cells, correlating with low proliferation and survival rates and poor production of Th1, Th2, and Th17 cytokines by memory CD4+ T cells. In naive CD4+ T cells, c-Rel is dispensable for early IL2 induction but contributes to later phases of IL2 expression. The patient's naive B cells displayed impaired MYC and BCL2L1 induction, compromising B cell survival and proliferation and preventing their differentiation into Ig-secreting plasmablasts. Inherited c-Rel deficiency disrupts the development and function of multiple myeloid and lymphoid cells, compromising innate and adaptive immunity to multiple infectious agents.


Asunto(s)
Genes rel , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/inmunología , Proteínas Proto-Oncogénicas c-rel/deficiencia , Proteínas Proto-Oncogénicas c-rel/genética , Inmunidad Adaptativa/genética , Inmunidad Adaptativa/inmunología , Niño , Consanguinidad , Femenino , Trasplante de Células Madre Hematopoyéticas , Homocigoto , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Activación de Linfocitos , Linfocitos/clasificación , Linfocitos/inmunología , Mutación , Células Mieloides/inmunología , Enfermedades de Inmunodeficiencia Primaria/terapia , Isoformas de Proteínas
8.
Science ; 373(6558)2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34446580

RESUMEN

The immune system has evolved in the face of microbial exposure. How maternal infection experienced at distinct developmental stages shapes the offspring immune system remains poorly understood. Here, we show that during pregnancy, maternally restricted infection can have permanent and tissue-specific impacts on offspring immunity. Mechanistically, maternal interleukin-6 produced in response to infection can directly impose epigenetic changes on fetal intestinal epithelial stem cells, leading to long-lasting impacts on intestinal immune homeostasis. As a result, offspring of previously infected dams develop enhanced protective immunity to gut infection and increased inflammation in the context of colitis. Thus, maternal infection can be coopted by the fetus to promote long-term, tissue-specific fitness, a phenomenon that may come at the cost of predisposition to inflammatory disorders.


Asunto(s)
Colitis/inmunología , Inmunidad , Interleucina-6/inmunología , Intestinos/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , Células Th17/inmunología , Infecciones por Yersinia pseudotuberculosis/inmunología , Animales , Candidiasis/inmunología , Cromatina/metabolismo , Epigénesis Genética , Epigenoma , Femenino , Desarrollo Fetal , Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/fisiología , Interleucina-6/sangre , Interleucina-6/farmacología , Mucosa Intestinal/citología , Mucosa Intestinal/embriología , Mucosa Intestinal/inmunología , Intestinos/embriología , Intestinos/microbiología , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal , Salmonelosis Animal/inmunología , Células Madre/inmunología , Células Madre/fisiología , Subgrupos de Linfocitos T/inmunología
9.
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
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(3): 254-256, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30742077
12.
Eur J Immunol ; 49(1): 11-18, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30350853

RESUMEN

Innate lymphoid cells (ILCs) represent a family of innate effector cells including NK cells, lymphoid tissue inducer (LTi) cells, and distinct ILC1, ILC2, and ILC3 subsets that produce IFN-γ, IL-5/IL-13, and IL-17A/IL-22, respectively. ILCs accumulate at mucosal sites and can promote the first-line defense against infection. ILCs are also implicated in tissue repair and can either pre-empt, or alternatively, exacerbate inflammation. Studies in mice have identified ILC precursors in fetal liver and adult BM that have diverse lineage potential. As such, these sites have been considered as the 'factories' to generate mature ILC. Here, we summarize knowledge concerning murine and human ILC development and discuss the recent identification of circulating multipotent and unipotent ILC precursors. We propose an alternative model of "ILC-poiesis", whereby blood ILC precursors migrate into tissues to complete their differentiation into mature ILC subsets under the influence of local environmental factors. Within this framework, ILC-poiesis guarantees appropriate ILC generation at the right place and the right time. We further discusss the potential applications of circulating ILC precursors for cell therapy of human disease.


Asunto(s)
Diferenciación Celular , Inmunidad Innata , Linfocitos/inmunología , Animales , Citocinas/metabolismo , Hematopoyesis , Humanos , Ratones
13.
Nat Methods ; 15(8): 623-630, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30065364

RESUMEN

Lymph nodes (LNs) facilitate the cellular interactions that orchestrate immune responses. Human immune system (HIS) mice are powerful tools for interrogation of human immunity but lack secondary lymphoid tissue (SLT) as a result of a deficiency in Il2rg-dependent lymphoid tissue inducer cells. To restore LN development, we induced expression of thymic-stromal-cell-derived lymphopoietin (TSLP) in a Balb/c Rag2-/-Il2rg-/-SirpaNOD (BRGS) HIS mouse model. The resulting BRGST HIS mice developed a full array of LNs with compartmentalized human B and T cells. Compared with BRGS HIS mice, BRGST HIS mice have a larger thymus, more mature B cells, and abundant IL-21-producing follicular helper T (TFH) cells, and show enhanced antigen-specific responses. Using BRGST HIS mice, we demonstrated that LN TFH cells are targets of acute HIV infection and represent a reservoir for latent HIV. In summary, BRGST HIS mice reflect the effects of SLT development on human immune responses and provide a model for visualization and interrogation of regulators of immunity.


Asunto(s)
Ganglios Linfáticos/crecimiento & desarrollo , Ganglios Linfáticos/inmunología , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Citocinas/genética , Citocinas/inmunología , Femenino , Infecciones por VIH/inmunología , VIH-1 , Humanos , Cambio de Clase de Inmunoglobulina , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Ganglios Linfáticos/citología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Inmunológicos , Linfocitos T/citología , Linfocitos T/inmunología , Latencia del Virus/inmunología , Linfopoyetina del Estroma Tímico
14.
Sci Immunol ; 3(24)2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29907691

RESUMEN

Heterozygosity for human signal transducer and activator of transcription 3 (STAT3) dominant-negative (DN) mutations underlies an autosomal dominant form of hyper-immunoglobulin E syndrome (HIES). We describe patients with an autosomal recessive form of HIES due to loss-of-function mutations of a previously uncharacterized gene, ZNF341 ZNF341 is a transcription factor that resides in the nucleus, where it binds a specific DNA motif present in various genes, including the STAT3 promoter. The patients' cells have low basal levels of STAT3 mRNA and protein. The autoinduction of STAT3 production, activation, and function by STAT3-activating cytokines is strongly impaired. Like patients with STAT3 DN mutations, ZNF341-deficient patients lack T helper 17 (TH17) cells, have an excess of TH2 cells, and have low memory B cells due to the tight dependence of STAT3 activity on ZNF341 in lymphocytes. Their milder extra-hematopoietic manifestations and stronger inflammatory responses reflect the lower ZNF341 dependence of STAT3 activity in other cell types. Human ZNF341 is essential for the STAT3 transcription-dependent autoinduction and sustained activity of STAT3.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Síndrome de Job/genética , Factor de Transcripción STAT3/genética , Factores de Transcripción/genética , Transcripción Genética/inmunología , Adolescente , Adulto , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Núcleo Celular/metabolismo , Consanguinidad , Citocinas/inmunología , Citocinas/metabolismo , Exones/genética , Femenino , Genes Recesivos/genética , Genes Recesivos/inmunología , Homocigoto , Humanos , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Síndrome de Job/sangre , Síndrome de Job/inmunología , Mutación con Pérdida de Función , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Linaje , Regiones Promotoras Genéticas/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT3/inmunología , Factor de Transcripción STAT3/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo , Secuenciación del Exoma , Adulto Joven , Dedos de Zinc/genética
15.
J Allergy Clin Immunol ; 142(6): 1793-1807, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29486229

RESUMEN

BACKGROUND: Group 2 innate lymphoid cells (ILC2s) are major producers of the cytokines driving allergic asthma, and increased ILC2 numbers have been detected in blood and sputum of asthmatic patients. Asthma susceptibility has a strong genetic component, but the underlying mechanisms and whether asthma genetics affect ILC2 biology remain unclear. OBJECTIVE: We sought to study the ILC2 transcriptome and epigenome during airway inflammation (AI) to couple these to genes and genetic variants associated with asthma pathogenesis. METHODS: Mice harboring a reporter for the key ILC2 transcription factor GATA-3 were subjected to IL-33-driven AI, and ILC2s were isolated from bronchoalveolar lavage fluid and mediastinal lymph nodes. Human ILC2s were purified from peripheral blood and activated in vitro. We used RNA sequencing, genome-wide identification of histone-3 lysine-4 dimethylation-marked chromatin, and computational approaches to study the ILC2 transcriptome and epigenome. RESULTS: Activated ILC2s in mice displayed a tissue-specific gene expression signature that emerged from remarkably similar epigenomes. We identified superenhancers implicated in controlling ILC2 identity and asthma-associated genes. More than 300 asthma-associated genetic polymorphisms identified in genome-wide association studies localized to H3K4Me2+ gene regulatory elements in ILC2s. A refined set of candidate causal asthma-associated variants was uniquely enriched in ILC2, but not TH2 cell, regulatory regions. CONCLUSIONS: ILC2s in AI use a flexible epigenome that couples adaptation to new microenvironments with functional plasticity. Importantly, we reveal strong correlations between gene regulatory mechanisms in ILC2s and the genetic basis of asthma, supporting a pathogenic role for ILC2s in patients with allergic asthma.


Asunto(s)
Asma/genética , Asma/inmunología , Predisposición Genética a la Enfermedad , Linfocitos/inmunología , Animales , Epigénesis Genética , Factor de Transcripción GATA3/genética , Genoma , Humanos , Inmunidad Innata , Ratones , Secuencias Reguladoras de Ácidos Nucleicos , Transcriptoma
16.
Nat Commun ; 8(1): 1762, 2017 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-29176694

RESUMEN

While patient selection and clinical management have reduced high-dose IL-2 (HDIL2) immunotherapy toxicities, the immune mechanisms that underlie HDIL2-induced morbidity remain unclear. Here we show that dose-dependent morbidity and mortality of IL-2 immunotherapy can be modeled in human immune system (HIS) mice. Depletion of human T cell subsets during the HDIL2 treatment reduces toxicity, pointing to the central function of T cells. Preferential expansion of effector T cells secondary to defective suppressive capacity of regulatory T (Treg) cells after HDIL2 therapy further underscores the importance of Treg in the maintenance of immune tolerance. IL-2 toxicity is induced by selective depletion or inhibition of Treg after LDIL2 therapy, and is ameliorated in HDIL2-treated HIS mice receiving the PIM-1 kinase inhibitor, Kaempferol. Modeling IL-2 pathophysiology in HIS mice offers a means to understand the functions of effector and regulatory T cells in immune-mediated toxicities associated with cancer immunotherapy.


Asunto(s)
Interleucina-2/inmunología , Neoplasias/terapia , Linfocitos T Reguladores/inmunología , Animales , Femenino , Humanos , Tolerancia Inmunológica , Inmunoterapia , Interleucina-2/administración & dosificación , Interleucina-2/genética , Interleucina-2/toxicidad , Ratones , Ratones Endogámicos BALB C , Neoplasias/inmunología , Linfocitos T Reguladores/efectos de los fármacos
17.
Oncotarget ; 8(47): 81729-81730, 2017 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-29137215
18.
Curr Opin Immunol ; 44: 61-68, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28359987

RESUMEN

Innate lymphoid cells (ILCs) are lineage- and antigen receptor-negative lymphocytes including natural killer (NK) cells and at least three distinguishable cell subsets (ILC1, ILC2, ILC3) that rapidly produce cytokines (IFN-γ, IL-5, IL-13, IL-17A, IL-22) upon activation. As such, ILCs can act as first-line defenders in the context of infection, inflammation and cancer. Because of the strong conservation between the expression of key transcription factors that can drive signature cytokine outputs in ILCs and differentiated helper T cells, it has been proposed that ILCs represent innate counterparts of the latter. Several distinct ILC precursors (ILCP) with pan-ILC (giving rise to all ILCs) or subset-restricted potentials have been described in both mouse and man. How and where these different ILCP give rise to more mature tissue-resident ILCs remains unclear. Recently, environmental signals have been shown to epigenetically influence canonical ILC differentiation pathways, generating substantial functional plasticity. These new results suggest that while ILC differentiation may be 'fixed' in principle, it remains 'flexible' in practice. A more comprehensive knowledge in the molecular mechanisms that regulate ILC development and effector functions may allow for therapeutic manipulation of ILCs for diverse disease conditions.


Asunto(s)
Inmunoterapia , Inflamación/inmunología , Linfocitos/fisiología , Células Progenitoras Linfoides/fisiología , Neoplasias/inmunología , Animales , Diferenciación Celular , Plasticidad de la Célula , Citocinas/metabolismo , Epigénesis Genética , Humanos , Inmunidad Innata , Activación de Linfocitos , Ratones , Linfocitos T Colaboradores-Inductores/fisiología
19.
Cell ; 168(6): 1086-1100.e10, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28283063

RESUMEN

Innate lymphoid cells (ILCs) represent innate versions of T helper and cytotoxic T cells that differentiate from committed ILC precursors (ILCPs). How ILCPs give rise to mature tissue-resident ILCs remains unclear. Here, we identify circulating and tissue ILCPs in humans that fail to express the transcription factors and cytokine outputs of mature ILCs but have these signature loci in an epigenetically poised configuration. Human ILCPs robustly generate all ILC subsets in vitro and in vivo. While human ILCPs express low levels of retinoic acid receptor (RAR)-related orphan receptor C (RORC) transcripts, these cells are found in RORC-deficient patients and retain potential for EOMES+ natural killer (NK) cells, interferon gamma-positive (IFN-γ+) ILC1s, interleukin (IL)-13+ ILC2s, and for IL-22+, but not for IL-17A+ ILC3s. Our results support a model of tissue ILC differentiation ("ILC-poiesis"), whereby diverse ILC subsets are generated in situ from systemically distributed ILCPs in response to local environmental signals.


Asunto(s)
Linfocitos/citología , Células Madre/citología , Animales , Antígenos CD34/análisis , Diferenciación Celular , Linaje de la Célula , Sangre Fetal/citología , Feto/citología , Humanos , Inmunidad Innata , Interleucina-17 , Hígado/citología , Pulmón/citología , Linfocitos/inmunología , Tejido Linfoide/citología , Ratones , Proteínas Proto-Oncogénicas c-kit/análisis , Transcripción Genética
20.
J Exp Med ; 213(4): 569-83, 2016 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-26976630

RESUMEN

Group 2 innate lymphoid cells (ILC2) include IL-5- and IL-13-producing CRTh2(+)CD127(+)cells that are implicated in early protective immunity at mucosal surfaces. Whereas functional plasticity has been demonstrated for both human and mouse ILC3 subsets that can reversibly give rise to IFN-γ-producing ILC1, plasticity of human or mouse ILC2 has not been shown. Here, we analyze the phenotypic and functional heterogeneity of human peripheral blood ILC2. Although subsets of human CRTh2(+)ILC2 differentially express CD117 (c-kit receptor), some ILC2 surface phenotypes are unstable and can be modulated in vitro. Surprisingly, human IL-13(+)ILC2 can acquire the capacity to produce IFN-γ, thereby generating plastic ILC2. ILC2 cultures demonstrated that IFN-γ(+)ILC2 clones could be derived and were stably associated with increased T-BET expression. The inductive mechanism for ILC2 plasticity was mapped to the IL-12-IL-12R signaling pathway and was confirmed through analysis of patients with Mendelian susceptibility to mycobacterial disease due to IL-12Rß1 deficiencies that failed to generate plastic ILC2. We also detected IL-13(+)IFN-γ(+)ILC2 ex vivo in intestinal samples from Crohn's disease patients. These results demonstrate cytokine production plasticity for human ILC2 and further suggest that environmental cues can dictate ILC phenotype and function for these tissue-resident innate effector cells.


Asunto(s)
Enfermedad de Crohn/inmunología , Inmunidad Innata , Interleucina-12/inmunología , Intestinos/inmunología , Linfocitos/inmunología , Animales , Células Cultivadas , Enfermedad de Crohn/patología , Femenino , Humanos , Interferón gamma/inmunología , Interleucina-13/inmunología , Interleucina-5/inmunología , Intestinos/patología , Linfocitos/patología , Masculino , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...