Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 22(2): 179-192, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33462452

RESUMEN

Metabolic programming controls immune cell lineages and functions, but little is known about γδ T cell metabolism. Here, we found that γδ T cell subsets making either interferon-γ (IFN-γ) or interleukin (IL)-17 have intrinsically distinct metabolic requirements. Whereas IFN-γ+ γδ T cells were almost exclusively dependent on glycolysis, IL-17+ γδ T cells strongly engaged oxidative metabolism, with increased mitochondrial mass and activity. These distinct metabolic signatures were surprisingly imprinted early during thymic development and were stably maintained in the periphery and within tumors. Moreover, pro-tumoral IL-17+ γδ T cells selectively showed high lipid uptake and intracellular lipid storage and were expanded in obesity and in tumors of obese mice. Conversely, glucose supplementation enhanced the antitumor functions of IFN-γ+ γδ T cells and reduced tumor growth upon adoptive transfer. These findings have important implications for the differentiation of effector γδ T cells and their manipulation in cancer immunotherapy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias del Colon/metabolismo , Metabolismo Energético , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Subgrupos de Linfocitos T/metabolismo , Timo/metabolismo , Microambiente Tumoral , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Linaje de la Célula , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Neoplasias del Colon/terapia , Femenino , Glucosa/metabolismo , Glucólisis , Humanos , Inmunoterapia Adoptiva , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Metabolismo de los Lípidos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/trasplante , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/metabolismo , Obesidad/inmunología , Obesidad/metabolismo , Técnicas de Cultivo de Órganos , Fenotipo , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/trasplante , Timo/inmunología , Carga Tumoral
2.
Nat Immunol ; 20(3): 373, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30728493

RESUMEN

In the version of this article initially published, three authors (Hui-Fern Kuoy, Adam P. Uldrich and Dale. I. Godfrey) and their affiliations, acknowledgments and contributions were not included. The correct information is as follows:Ayano C. Kohlgruber1,2, Shani T. Gal-Oz3, Nelson M. LaMarche1,2, Moto Shimazaki1, Danielle Duquette4, Hui-Fern Koay5,6, Hung N. Nguyen1, Amir I. Mina4, Tyler Paras1, Ali Tavakkoli7, Ulrich von Andrian2,8, Adam P. Uldrich5,6, Dale I. Godfrey5,6, Alexander S. Banks4, Tal Shay3, Michael B. Brenner1,10* and Lydia Lynch1,4,9,10*1Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA, USA. 2Division of Medical Sciences, Harvard Medical School, Boston, MA, USA. 3Department of Life Sciences, Ben-Gurion University of the Negev, Beersheba, Israel. 4Division of Endocrinology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA. 5Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Australia. 6ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Australia. 7Department of General and Gastrointestinal Surgery, Brigham and Women's Hospital, Boston, MA, USA. 8Department of Microbiology and Immunology, Harvard Medical School, Boston, MA, USA. 9School of Biochemistry and Immunology, Trinity College, Dublin, Ireland. 10These authors jointly supervised this work: Michael B. Brenner, Lydia Lynch. *e-mail: mbrenner@research.bwh.harvard.edu; llynch@bwh.harvard.eduAcknowledgementsWe thank A.T. Chicoine, flow cytometry core manager at the Human Immunology Center at BWH, for flow cytometry sorting. We thank D. Sant'Angelo (Rutgers Cancer Institute) for providing Zbtb16-/- mice and R. O'Brien (National Jewish Health) for providing Vg4/6-/- mice. Supported by NIH grant R01 AI11304603 (to M.B.B.), ERC Starting Grant 679173 (to L.L.), the National Health and Medical Research Council of Australia (1013667), an Australian Research Council Future Fellowship (FT140100278 for A.P.U.) and a National Health and Medical Research Council of Australia Senior Principal Research Fellowship (1117766 for D.I.G.).Author contributionsA.C.K., L.L., and M.B.B. conceived and designed the experiments, and wrote the manuscript. A.C.K., N.M.L., L.L., H.N.N., M.S., T.P., and D.D. performed the experiments. S.T.G.-O. and T.S. performed the RNA-seq analysis. A.S.B. and A.I.M. provided advice and performed the CLAMS experiments. A.T. provided human bariatric patient samples. Parabiosis experiments were performed in the laboratory of U.v.A. H.-F.K., A.P.U. and D.I.G provided critical insight into the TCR chain usage of PLZF+ γδ T cells. M.B.B., N.M.L., and L.L. critically reviewed the manuscript.The errors have been corrected in the HTML and PDF version of the article.Correction to: Nature Immunology doi:10.1038/s41590-018-0094-2 (2018), published online 18 April 2018.

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.
Nat Immunol ; 19(5): 464-474, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29670241

RESUMEN

γδ T cells are situated at barrier sites and guard the body from infection and damage. However, little is known about their roles outside of host defense in nonbarrier tissues. Here, we characterize a highly enriched tissue-resident population of γδ T cells in adipose tissue that regulate age-dependent regulatory T cell (Treg) expansion and control core body temperature in response to environmental fluctuations. Mechanistically, innate PLZF+ γδ T cells produced tumor necrosis factor and interleukin (IL) 17 A and determined PDGFRα+ and Pdpn+ stromal-cell production of IL-33 in adipose tissue. Mice lacking γδ T cells or IL-17A exhibited decreases in both ST2+ Treg cells and IL-33 abundance in visceral adipose tissue. Remarkably, these mice also lacked the ability to regulate core body temperature at thermoneutrality and after cold challenge. Together, these findings uncover important physiological roles for resident γδ T cells in adipose tissue immune homeostasis and body-temperature control.


Asunto(s)
Tejido Adiposo/citología , Homeostasis/fisiología , Interleucina-17/metabolismo , Linfocitos T Reguladores/fisiología , Termogénesis/fisiología , Tejido Adiposo/fisiología , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Antígenos de Linfocitos T gamma-delta , Subgrupos de Linfocitos T/fisiología
5.
Nat Immunol ; 18(11): 1197-1206, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28920951

RESUMEN

Activated natural killer (NK) cells engage in a robust metabolic response that is required for normal effector function. Using genetic, pharmacological and metabolic analyses, we demonstrated an essential role for Srebp transcription factors in cytokine-induced metabolic reprogramming of NK cells that was independent of their conventional role in the control of lipid synthesis. Srebp was required for elevated glycolysis and oxidative phosphorylation and promoted a distinct metabolic pathway configuration in which glucose was metabolized to cytosolic citrate via the citrate-malate shuttle. Preventing the activation of Srebp or direct inhibition of the citrate-malate shuttle inhibited production of interferon-γ and NK cell cytotoxicity. Thus, Srebp controls glucose metabolism in NK cells, and this Srebp-dependent regulation is critical for NK cell effector function.


Asunto(s)
Glucosa/metabolismo , Glucólisis , Células Asesinas Naturales/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Animales , Proliferación Celular , Citocinas/metabolismo , Citometría de Flujo , Humanos , Immunoblotting , Células Asesinas Naturales/inmunología , Lípidos/biosíntesis , Fosforilación Oxidativa , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
6.
Nat Immunol ; 16(1): 85-95, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25436972

RESUMEN

Invariant natural killer T cells (iNKT cells) are lipid-sensing innate T cells that are restricted by the antigen-presenting molecule CD1d and express the transcription factor PLZF. iNKT cells accumulate in adipose tissue, where they are anti-inflammatory, but the factors that contribute to their anti-inflammatory nature, as well as their targets in adipose tissue, are unknown. Here we found that iNKT cells in adipose tissue had a unique transcriptional program and produced interleukin 2 (IL-2) and IL-10. Unlike other iNKT cells, they lacked PLZF but expressed the transcription factor E4BP4, which controlled their IL-10 production. The adipose iNKT cells were a tissue-resident population that induced an anti-inflammatory phenotype in macrophages and, through the production of IL-2, controlled the number, proliferation and suppressor function of regulatory T cells (Treg cells) in adipose tissue. Thus, iNKT cells in adipose tissue are unique regulators of immunological homeostasis in this tissue.


Asunto(s)
Tejido Adiposo/inmunología , Factores de Transcripción de Tipo Kruppel/biosíntesis , Macrófagos/inmunología , Células T Asesinas Naturales/metabolismo , Linfocitos T Reguladores/metabolismo , Tejido Adiposo/citología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/inmunología , Procesos de Crecimiento Celular/inmunología , Femenino , Citometría de Flujo , Regulación de la Expresión Génica , Homeostasis/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-2/genética , Interleucina-2/inmunología , Factores de Transcripción de Tipo Kruppel/deficiencia , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/inmunología , Macrófagos/citología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/citología , Células T Asesinas Naturales/inmunología , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Organismos Libres de Patógenos Específicos , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología
7.
Immunity ; 48(1): 9-11, 2018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29343443

RESUMEN

NK cells control tumor growth directly through targeted cytotoxic granule release or cytokine secretion and indirectly by orchestrating anti-tumor immune responses. In this issue of Immunity, Glasner et al. (2018) now reveal a new role for NK cells in preventing metastatic spread through controlling tumor architecture.


Asunto(s)
Células Asesinas Naturales , Microambiente Tumoral , Gránulos Citoplasmáticos , Humanos , Neoplasias
8.
Trends Immunol ; 44(3): 159-161, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36754745

RESUMEN

Tumors can evade conventional T cell recognition by rendering the HLA class I antigen presentation system defective. In a recent study, de Vries et al. reveal γδ T cells as key contributors to the efficacy of immune checkpoint blockade (ICB) against HLA-I-silenced cancers, highlighting a novel layer of surveillance against immune escape by tumors.


Asunto(s)
Neoplasias , Humanos , Antígenos de Histocompatibilidad Clase I , Linfocitos T
9.
Immunity ; 46(2): 273-286, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28228283

RESUMEN

Adipose tissue has a dynamic immune system that adapts to changes in diet and maintains homeostatic tissue remodeling. Adipose type 1 innate lymphoid cells (AT1-ILCs) promote pro-inflammatory macrophages in obesity, but little is known about their functions at steady state. Here we found that human and murine adipose tissue harbor heterogeneous populations of AT1-ILCs. Experiments using parabiotic mice fed a high-fat diet (HFD) showed differential trafficking of AT1-ILCs, particularly in response to short- and long-term HFD and diet restriction. At steady state, AT1-ILCs displayed cytotoxic activity toward adipose tissue macrophages (ATMs). Depletion of AT1-ILCs and perforin deficiency resulted in alterations in the ratio of inflammatory to anti-inflammatory ATMs, and adoptive transfer of AT1-ILCs exacerbated metabolic disorder. Diet-induced obesity impaired AT1-ILC killing ability. Our findings reveal a role for AT1-ILCs in regulating ATM homeostasis through cytotoxicity and suggest that this function is relevant in both homeostasis and metabolic disease.


Asunto(s)
Tejido Adiposo/inmunología , Citotoxicidad Inmunológica/inmunología , Homeostasis/inmunología , Linfocitos/inmunología , Macrófagos/inmunología , Obesidad/inmunología , Tejido Adiposo/citología , Animales , Femenino , Humanos , Inmunidad Innata , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/patología
10.
J Immunol ; 211(4): 633-647, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37449888

RESUMEN

NK cells and CD8 T cells use cytotoxic molecules to kill virally infected and tumor cell targets. While perforin and granzyme B (GzmB) are the most commonly studied lytic molecules, less is known about granzyme K (GzmK). However, this granzyme has been recently associated with improved prognosis in solid tumors. In this study, we show that, in humans, GzmK is predominantly expressed by innate-like lymphocytes, as well as a newly identified population of GzmK+CD8+ non- mucosal-associated invariant T cells with innate-like characteristics. We found that GzmK+ T cells are KLRG1+EOMES+IL-7R+CD62L-Tcf7int, suggesting that they are central memory T and effector memory T cells. Furthermore, GzmK+ cells are absent/low in cord blood, suggesting that GzmK is upregulated with immune experience. Surprisingly, GzmK+ cells respond to cytokine stimuli alone, whereas TCR stimulation downregulates GzmK expression, coinciding with GzmB upregulation. GzmK+ cells have reduced IFN-γ production compared with GzmB+ cells in each T cell lineage. Collectively, this suggests that GzmK+ cells are not naive, and they may be an intermediate memory-like or preterminally differentiated population. GzmK+ cells are enriched in nonlymphoid tissues such as the liver and adipose. In colorectal cancer, GzmK+ cells are enriched in the tumor and can produce IFN-γ, but GzmK+ expression is mutually exclusive with IL-17a production. Thus, in humans, GzmK+ cells are innate memory-like cells that respond to cytokine stimulation alone and may be important effector cells in the tumor.


Asunto(s)
Linfocitos T CD8-positivos , Citocinas , Granzimas , Humanos , Citocinas/metabolismo , Granzimas/metabolismo , Células Asesinas Naturales , Receptores de Antígenos de Linfocitos T/metabolismo
11.
J Immunol ; 208(6): 1445-1455, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35181637

RESUMEN

Plasmacytoid dendritic cells (pDCs) display an increased abundance in visceral adipose tissue (VAT) of humans with obesity. In the current study, we set out to decipher the molecular mechanisms of their recruitment to VAT and the functional relevance of this process. We observed increased pDC numbers in murine blood, liver, spleen, and VAT after feeding a high-fat diet (HFD) for 3 wk when compared with a standard diet. pDCs were enriched in fat-associated lymphoid clusters representing highly specific lymphoid regions within VAT. HFD led to an enlargement of fat-associated lymphoid clusters with an increased density and migratory speed of pDCs as shown by intravital multiphoton microscopy. For their recruitment into VAT, pDCs employed P-selectin with E-selectin and L-selectin being only critical in response to HFD, indicating that the molecular cues underlying pDC trafficking were dependent on the nutritional state. Subsequent recruitment steps required α4ß1 and α4ß7 integrins and engagement of CCR7. Application of fingolimod (FTY720) abrogated egress of pDCs from VAT, indicating the involvement of sphingosine-1-phosphate in this process. Furthermore, HFD altered pDC functions by promoting their activation and type 1 IFN expression. Blocking pDC infiltration into VAT prevented weight gain and improved glucose tolerance during HFD. In summary, a HFD fundamentally alters pDC biology by promoting their trafficking, retention, and activation in VAT, which in turn seems to regulate metabolism.


Asunto(s)
Dieta Alta en Grasa , Grasa Intraabdominal , Tejido Adiposo , Animales , Células Dendríticas , Grasa Intraabdominal/metabolismo , Ratones , Fenotipo
12.
Nature ; 559(7715): 637-641, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30022161

RESUMEN

Diabetes is a complex metabolic syndrome that is characterized by prolonged high blood glucose levels and frequently associated with life-threatening complications1,2. Epidemiological studies have suggested that diabetes is also linked to an increased risk of cancer3-5. High glucose levels may be a prevailing factor that contributes to the link between diabetes and cancer, but little is known about the molecular basis of this link and how the high glucose state may drive genetic and/or epigenetic alterations that result in a cancer phenotype. Here we show that hyperglycaemic conditions have an adverse effect on the DNA 5-hydroxymethylome. We identify the tumour suppressor TET2 as a substrate of the AMP-activated kinase (AMPK), which phosphorylates TET2 at serine 99, thereby stabilizing the tumour suppressor. Increased glucose levels impede AMPK-mediated phosphorylation at serine 99, which results in the destabilization of TET2 followed by dysregulation of both 5-hydroxymethylcytosine (5hmC) and the tumour suppressive function of TET2 in vitro and in vivo. Treatment with the anti-diabetic drug metformin protects AMPK-mediated phosphorylation of serine 99, thereby increasing TET2 stability and 5hmC levels. These findings define a novel 'phospho-switch' that regulates TET2 stability and a regulatory pathway that links glucose and AMPK to TET2 and 5hmC, which connects diabetes to cancer. Our data also unravel an epigenetic pathway by which metformin mediates tumour suppression. Thus, this study presents a new model for how a pernicious environment can directly reprogram the epigenome towards an oncogenic state, offering a potential strategy for cancer prevention and treatment.


Asunto(s)
Adenilato Quinasa/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Diabetes Mellitus/metabolismo , Glucosa/metabolismo , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/metabolismo , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Animales , ADN/química , ADN/metabolismo , Metilación de ADN , Diabetes Mellitus/genética , Dioxigenasas , Estabilidad de Enzimas , Epigénesis Genética , Hemoglobina Glucada/análisis , Humanos , Hiperglucemia/metabolismo , Metformina/farmacología , Metformina/uso terapéutico , Ratones , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Fosforilación , Fosfoserina/metabolismo , Especificidad por Sustrato , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Int J Cancer ; 153(1): 120-132, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-36883413

RESUMEN

Resistance to platinum-based chemotherapy is the major cause of death from high-grade serous ovarian cancer (HGSOC). We hypothesise that detection of specific DNA methylation changes may predict platinum resistance in HGSOC. Using a publicly available "discovery" dataset we examined epigenomic and transcriptomic alterations between primary platinum-sensitive (n = 32) and recurrent acquired drug resistant HGSOC (n = 28) and identified several genes involved in immune and chemoresistance-related pathways. Validation via high-resolution melt analysis of these findings, in cell lines and HGSOC tumours, demonstrated the most consistent changes were observed in three of the genes: APOBEC3A, NKAPL and PDCD1. Plasma samples from an independent HGSOC cohort (n = 17) were analysed using droplet digital PCR. Hypermethylation of NKAPL was detected in 46% and hypomethylation of APOBEC3A in 69% of plasma samples taken from women with relapsed HGSOC (n = 13), with no alterations identified in disease-free patients (n = 4). Following these results, and using a CRISPR-Cas9 approach, we were also able to demonstrate that in vitro NKAPL promoter demethylation increased platinum sensitivity by 15%. Overall, this study demonstrates the importance of aberrant methylation, especially of the NKAPL gene, in acquired platinum resistance in HGSOC.


Asunto(s)
Cistadenocarcinoma Seroso , Neoplasias Ováricas , Humanos , Femenino , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Platino (Metal)/farmacología , Platino (Metal)/uso terapéutico , Resistencia a Antineoplásicos/genética , Epigenómica , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Carcinoma Epitelial de Ovario , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patología
14.
EMBO J ; 38(14): e101260, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31304630

RESUMEN

Tissue-resident iNKT cells maintain tissue homeostasis and peripheral surveillance against pathogens; however, studying these cells is challenging due to their low abundance and poor recovery from tissues. We here show that iNKT transnuclear mice, generated by somatic cell nuclear transfer, have increased tissue resident iNKT cells. We examined expression of PLZF, T-bet, and RORγt, as well as cytokine/chemokine profiles, and found that both monoclonal and polyclonal iNKT cells differentiated into functional subsets that faithfully replicated those seen in wild-type mice. We detected iNKT cells from tissues in which they are rare, including adipose, lung, skin-draining lymph nodes, and a previously undescribed population in Peyer's patches (PP). PP-NKT cells produce the majority of the IL-4 in Peyer's patches and provide indirect help for B-cell class switching to IgG1 in both transnuclear and wild-type mice. Oral vaccination with α-galactosylceramide shows enhanced fecal IgG1 titers in iNKT cell-sufficient mice. Transcriptional profiling reveals a unique signature of PP-NKT cells, characterized by tissue residency. We thus define PP-NKT as potentially important for surveillance for mucosal pathogens.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Cambio de Clase de Inmunoglobulina , Inmunoglobulina G/genética , Células T Asesinas Naturales/metabolismo , Ganglios Linfáticos Agregados/inmunología , Animales , Diferenciación Celular , Células Cultivadas , Femenino , Galactosilceramidas/administración & dosificación , Galactosilceramidas/inmunología , Interleucina-4/genética , Ratones , Células T Asesinas Naturales/citología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Técnicas de Transferencia Nuclear , Proteína de la Leucemia Promielocítica con Dedos de Zinc/genética , Proteínas de Dominio T Box/genética , Vacunación
15.
Cancer Immunol Immunother ; 71(12): 2943-2955, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35523889

RESUMEN

Invariant natural killer T cells (iNKT cells) express a semi-invariant T cell receptor that recognizes certain glycolipids (including α-galactosylceramide, αGC) bound to CD1d, and can induce potent antitumor responses. Here, we assessed whether αGC could enhance the efficacy of a GM-CSF-producing tumor cell vaccine in the transgenic SV40 T antigen-driven TRAMP prostate cancer model. In healthy mice, we initially found that optimal T cell responses were obtained with αGC-pulsed TRAMP-C2 cells secreting GM-CSF and milk fat globule epidermal growth factor protein-8 (MFG-E8) with an RGD to RGE mutation (GM-CSF/RGE TRAMP-C2), combined with systemic low dose IL-12. In a therapeutic model, transgenic TRAMP mice were then castrated at ~ 20 weeks, followed by treatment with the combination vaccine. Untreated mice succumbed to tumor by ~ 40 weeks, but survival was markedly prolonged by vaccine treatment, with most mice surviving past 80 weeks. Prostates in the treated mice were heavily infiltrated with T cells and iNKT cells, which both secreted IFNγ in response to tumor cells. The vaccine was not effective if the αGC, IL-12, or GM-CSF secretion was eliminated. Finally, immunized mice were fully resistant to challenge with TRAMP-C2 cells. Together these findings support further development of therapeutic vaccines that exploit iNKT cell activation.


Asunto(s)
Vacunas contra el Cáncer , Células T Asesinas Naturales , Neoplasias de la Próstata , Masculino , Ratones , Animales , Humanos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Activación de Linfocitos , Galactosilceramidas , Interleucina-12/farmacología , Neoplasias de la Próstata/terapia , Neoplasias de la Próstata/metabolismo , Vacunas Combinadas/farmacología , Antígenos Virales de Tumores , Familia de Proteínas EGF/metabolismo , Familia de Proteínas EGF/farmacología , Oligopéptidos/farmacología , Ratones Endogámicos C57BL
16.
Trends Immunol ; 40(9): 857-872, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31399336

RESUMEN

Adipose tissue is a critical regulator of systemic metabolism and normal bodily homeostasis. Recently, the immune system has been implicated in the regulation of adipose tissue homeostasis and function. Here, we highlight new insights into the distinct phenotypes and functions of adipose resident leukocytes, including a growing body of work on their role in adipose tissue remodeling and thermogenesis. We discuss how the protective capacity of the adipose immune system can be diminished during obesity, where immune cells promote sterile inflammation leading to insulin resistance. Understanding the role of adipose immune cells across different physiological states and processes is important in understanding the full breadth of adipose immunity and the possibilities to harness immune cells in fat tissue for the treatment of chronic metabolic conditions, including obesity.


Asunto(s)
Tejido Adiposo/inmunología , Homeostasis/inmunología , Inmunidad Innata/inmunología , Animales , Humanos
17.
Br J Cancer ; 125(10): 1341-1349, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34302062

RESUMEN

Colorectal cancer represents the second leading cause of cancer-related death worldwide. The therapeutic field of immuno-oncology has rapidly gained momentum, with strikingly promising results observed in clinical practice. Increasing emphasis has been placed on the role of the immune response in tumorigenesis, therapy and predicting prognosis. Enhanced understanding of the dynamic and complex tumour-immune microenvironment has enabled the development of molecularly directed, individualised treatment. Analysis of intra-tumoural lymphocyte infiltration and the dichotomisation of colorectal cancer into microsatellite stable and unstable disease has important therapeutic and prognostic implications, with potential to capitalise further on this data. This review discusses the latest evidence surrounding the tumour biology and immune landscape of colorectal cancer, novel immunotherapies and the interaction of the immune system with each apex of the tripartite of cancer management (oncotherapeutics, radiotherapy and surgery). By utilising the synergy of chemotherapeutic agents and immunotherapies, and identifying prognostic and predictive immunological biomarkers, we may enter an era of unprecedented disease control, survivorship and cure rates.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Neoplasias Colorrectales/inmunología , Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Humanos , Inmunoterapia , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Medicina de Precisión , Pronóstico , Microambiente Tumoral/efectos de los fármacos
18.
Immunity ; 37(3): 574-87, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22981538

RESUMEN

Invariant natural killer T (iNKT) cells are evolutionarily conserved innate T cells that influence inflammatory responses. We have shown that iNKT cells, previously thought to be rare in humans, were highly enriched in human and murine adipose tissue, and that as adipose tissue expanded in obesity, iNKT cells were depleted, correlating with proinflammatory macrophage infiltration. iNKT cell numbers were restored in mice and humans after weight loss. Mice lacking iNKT cells had enhanced weight gain, larger adipocytes, fatty livers, and insulin resistance on a high-fat diet. Adoptive transfer of iNKT cells into obese mice or in vivo activation of iNKT cells via their lipid ligand, alpha-galactocylceramide, decreased body fat, triglyceride levels, leptin, and fatty liver and improved insulin sensitivity through anti-inflammatory cytokine production by adipose-derived iNKT cells. This finding highlights the potential of iNKT cell-targeted therapies, previously proven to be safe in humans, in the management of obesity and its consequences.


Asunto(s)
Tejido Adiposo/inmunología , Citocinas/inmunología , Enfermedades Metabólicas/inmunología , Células T Asesinas Naturales/inmunología , Obesidad/inmunología , Tejido Adiposo/metabolismo , Traslado Adoptivo , Adulto , Animales , Antígenos CD1d/genética , Antígenos CD1d/inmunología , Antígenos CD1d/metabolismo , Antígeno CD11c/inmunología , Antígeno CD11c/metabolismo , Citocinas/metabolismo , Dieta Alta en Grasa/efectos adversos , Femenino , Citometría de Flujo , Humanos , Hígado/inmunología , Hígado/metabolismo , Recuento de Linfocitos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Persona de Mediana Edad , Células T Asesinas Naturales/metabolismo , Células T Asesinas Naturales/trasplante , Obesidad/etiología , Obesidad/metabolismo , Bazo/inmunología , Bazo/metabolismo , Adulto Joven
19.
J Immunol ; 202(12): 3404-3411, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31076528

RESUMEN

Obesity underpins the development of numerous chronic diseases, such as type II diabetes mellitus. It is well established that obesity negatively alters immune cell frequencies and functions. Mucosal-associated invariant T (MAIT) cells are a population of innate T cells, which we have previously reported are dysregulated in obesity, with altered circulating and adipose tissue frequencies and a reduction in their IFN-γ production, which is a critical effector function of MAIT cells in host defense. Hence, there is increased urgency to characterize the key molecular mechanisms that drive MAIT cell effector functions and to identify those which are impaired in the obesity setting. In this study, we found that MAIT cells significantly upregulate their rates of glycolysis upon activation in an mTORC1-dependent manner, and this is essential for MAIT cell IFN-γ production. Furthermore, we show that mTORC1 activation is dependent on amino acid transport via SLC7A5. In obese patients, using RNA sequencing, Seahorse analysis, and a series of in vitro experiments, we demonstrate that MAIT cells isolated from obese adults display defective glycolytic metabolism, mTORC1 signaling, and SLC7A5 aa transport. Collectively, our data detail the intrinsic metabolic pathways controlling MAIT cell cytokine production and highlight mTORC1 as an important metabolic regulator that is impaired in obesity, leading to altered MAIT cell responses.


Asunto(s)
Diabetes Mellitus Tipo 2/inmunología , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Células T Invariantes Asociadas a Mucosa/fisiología , Obesidad/inmunología , Adulto , Células Cultivadas , Femenino , Glucólisis , Humanos , Interferón gamma/metabolismo , Activación de Linfocitos , Masculino , Análisis de Secuencia de ARN , Transducción de Señal
20.
Semin Immunol ; 28(5): 431-440, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27789087

RESUMEN

At the simplest interpretation of the word, Immunometabolism describes the intersection of the fields of immunology and metabolism. With rapidly growing interest in this field, the term has expanded, and now encompasses a variety of concepts and definitions shaped by an individual's scientific area of expertise, cell-type and tissue of interest, and biological approach. One scientist may be interested in investigating the intrinsic metabolic checkpoints that drive a M1 versus M2 macrophage response, while another may be interested in how macrophages affect systemic metabolism during obesity. Although both interests have very different foci, they both reflect the current interests in immunometabolism and studies over the last decade have uncovered new metabolic nodes that dictate the course of effector fate within cells, as well as an unexpected role for the immune system in controlling systemic metabolism. Thus, immunometabolism is at the frontier for many novel therapeutic targets to control both cell intrinsic and whole body metabolism in many diseases including cancer, diabetes, obesity, and sepsis among others. In this review, we hope to break down the word immunometabolism into two main themes: whole-body metabolism and cellular bioenergetics. In each instance we will focus on the adipose tissue and its resident immune cells to illustrate recent advances in both sectors of immunometabolism.


Asunto(s)
Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Metabolismo Energético , Inmunidad , Inmunomodulación , Animales , Susceptibilidad a Enfermedades , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Redes y Vías Metabólicas , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA