Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(9): 2117-2119, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38670068

RESUMEN

While some people pore over the textbook and train through the classics of the field, many scientists come to immunology when they discover it intersecting with their "first love" interests. Five of these "accidental immunologists" tell us how they found their way to a fascination with the immune system.


Asunto(s)
Alergia e Inmunología , Humanos , Historia del Siglo XX , Historia del Siglo XXI , Animales , Sistema Inmunológico
2.
Annu Rev Cell Dev Biol ; 37: 549-573, 2021 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-34613819

RESUMEN

The prevalence of obesity is on the rise. What was once considered a simple disease of energy imbalance is now recognized as a complex condition perpetuated by neuro- and immunopathologies. In this review, we summarize the current knowledge of the neuroimmunoendocrine mechanisms underlying obesity. We examine the pleiotropic effects of leptin action in addition to its established role in the modulation of appetite, and we discuss the neural circuitry mediating leptin action and how this is altered with obesity, both centrally (leptin resistance) and in adipose tissues (sympathetic neuropathy). Finally, we dissect the numerous causal and consequential roles of adipose tissue macrophages in obesity and highlight recent key studies demonstrating their direct role in organismal energy homeostasis.


Asunto(s)
Tejido Adiposo , Obesidad , Homeostasis , Humanos , Obesidad/genética
3.
Immunity ; 57(1): 141-152.e5, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38091996

RESUMEN

Adipose tissues (ATs) are innervated by sympathetic nerves, which drive reduction of fat mass via lipolysis and thermogenesis. Here, we report a population of immunomodulatory leptin receptor-positive (LepR+) sympathetic perineurial barrier cells (SPCs) present in mice and humans, which uniquely co-express Lepr and interleukin-33 (Il33) and ensheath AT sympathetic axon bundles. Brown ATs (BATs) of mice lacking IL-33 in SPCs (SPCΔIl33) had fewer regulatory T (Treg) cells and eosinophils, resulting in increased BAT inflammation. SPCΔIl33 mice were more susceptible to diet-induced obesity, independently of food intake. Furthermore, SPCΔIl33 mice had impaired adaptive thermogenesis and were unresponsive to leptin-induced rescue of metabolic adaptation. We therefore identify LepR+ SPCs as a source of IL-33, which orchestrate an anti-inflammatory BAT environment, preserving sympathetic-mediated thermogenesis and body weight homeostasis. LepR+IL-33+ SPCs provide a cellular link between leptin and immune regulation of body weight, unifying neuroendocrinology and immunometabolism as previously disconnected fields of obesity research.


Asunto(s)
Tejido Adiposo Pardo , Leptina , Animales , Humanos , Ratones , Tejido Adiposo Pardo/inervación , Tejido Adiposo Pardo/metabolismo , Peso Corporal , Metabolismo Energético/fisiología , Interleucina-33/genética , Interleucina-33/metabolismo , Obesidad/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Termogénesis/fisiología
4.
Immunity ; 56(4): 747-749, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-37044065

RESUMEN

Fasting is known to impact monocyte dynamics and phenotype, but the mechanics and functional significance of this response remain unclear. In this issue of Immunity, Janssen and colleagues demonstrate that fasting and re-feeding causes monocytes to re-enter the bone marrow and alter the host response to infection.


Asunto(s)
Médula Ósea , Monocitos , Células de la Médula Ósea
5.
Cell ; 163(2): 381-93, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26411290

RESUMEN

RORγt(+) Th17 cells are important for mucosal defenses but also contribute to autoimmune disease. They accumulate in the intestine in response to microbiota and produce IL-17 cytokines. Segmented filamentous bacteria (SFB) are Th17-inducing commensals that potentiate autoimmunity in mice. RORγt(+) T cells were induced in mesenteric lymph nodes early after SFB colonization and distributed across different segments of the gastrointestinal tract. However, robust IL-17A production was restricted to the ileum, where SFB makes direct contact with the epithelium and induces serum amyloid A proteins 1 and 2 (SAA1/2), which promote local IL-17A expression in RORγt(+) T cells. We identified an SFB-dependent role of type 3 innate lymphoid cells (ILC3), which secreted IL-22 that induced epithelial SAA production in a Stat3-dependent manner. This highlights the critical role of tissue microenvironment in activating effector functions of committed Th17 cells, which may have important implications for how these cells contribute to inflammatory disease.


Asunto(s)
Microbioma Gastrointestinal , Interleucinas/metabolismo , Intestinos/inmunología , Receptores de Interleucina/metabolismo , Proteína Amiloide A Sérica/metabolismo , Células Th17/inmunología , Animales , Inmunidad Innata , Interleucinas/inmunología , Intestinos/anatomía & histología , Intestinos/microbiología , Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina/inmunología , Transducción de Señal , Interleucina-22
6.
Cell ; 163(1): 84-94, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26406372

RESUMEN

Leptin is a hormone produced by the adipose tissue that acts in the brain, stimulating white fat breakdown. We find that the lipolytic effect of leptin is mediated through the action of sympathetic nerve fibers that innervate the adipose tissue. Using intravital two-photon microscopy, we observe that sympathetic nerve fibers establish neuro-adipose junctions, directly "enveloping" adipocytes. Local optogenetic stimulation of sympathetic inputs induces a local lipolytic response and depletion of white adipose mass. Conversely, genetic ablation of sympathetic inputs onto fat pads blocks leptin-stimulated phosphorylation of hormone-sensitive lipase and consequent lipolysis, as do knockouts of dopamine ß-hydroxylase, an enzyme required for catecholamine synthesis. Thus, neuro-adipose junctions are necessary and sufficient for the induction of lipolysis in white adipose tissue and are an efferent effector of leptin action. Direct activation of sympathetic inputs to adipose tissues may represent an alternative approach to induce fat loss, circumventing central leptin resistance. PAPERCLIP.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Leptina/metabolismo , Lipólisis , Tejido Adiposo Blanco/inervación , Animales , Humanos , Ratones , Fosforilación , Receptores Adrenérgicos beta/metabolismo , Sistema Nervioso Simpático/metabolismo
7.
Immunity ; 52(6): 900-902, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32553178

RESUMEN

The hypothalamic-pituitary-adrenal axis modulates immunity in response to stress. In a recent report in the May 14, 2020 issue of Nature, Zhang et al. use optogenetic tools to investigate whether the splenic immune response is directly controlled by descending neuronal circuits activated in response to stress.


Asunto(s)
Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal , Encéfalo , Inmunidad Humoral , Neuronas
8.
Nat Immunol ; 21(2): 106-108, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31900463
9.
Nature ; 597(7876): 410-414, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34408322

RESUMEN

Signals from sympathetic neurons and immune cells regulate adipocytes and thereby contribute to fat tissue biology. Interactions between the nervous and immune systems have recently emerged as important regulators of host defence and inflammation1-4. Nevertheless, it is unclear whether neuronal and immune cells co-operate in brain-body axes to orchestrate metabolism and obesity. Here we describe a neuro-mesenchymal unit that controls group 2 innate lymphoid cells (ILC2s), adipose tissue physiology, metabolism and obesity via a brain-adipose circuit. We found that sympathetic nerve terminals act on neighbouring adipose mesenchymal cells via the ß2-adrenergic receptor to control the expression of glial-derived neurotrophic factor (GDNF) and the activity of ILC2s in gonadal fat. Accordingly, ILC2-autonomous manipulation of the GDNF receptor machinery led to alterations in ILC2 function, energy expenditure, insulin resistance and propensity to obesity. Retrograde tracing and chemical, surgical and chemogenetic manipulations identified a sympathetic aorticorenal circuit that modulates ILC2s in gonadal fat and connects to higher-order brain areas, including the paraventricular nucleus of the hypothalamus. Our results identify a neuro-mesenchymal unit that translates cues from long-range neuronal circuitry into adipose-resident ILC2 function, thereby shaping host metabolism and obesity.


Asunto(s)
Tejido Adiposo/inervación , Tejido Adiposo/metabolismo , Encéfalo/metabolismo , Inmunidad Innata/inmunología , Mesodermo/citología , Vías Nerviosas , Neuronas/citología , Obesidad/metabolismo , Tejido Adiposo/citología , Animales , Encéfalo/citología , Señales (Psicología) , Citocinas/metabolismo , Metabolismo Energético , Femenino , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Gónadas/metabolismo , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Proteínas Proto-Oncogénicas c-ret/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Sistema Nervioso Simpático/citología , Sistema Nervioso Simpático/metabolismo
11.
Nature ; 562(7725): 150, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29973715

RESUMEN

Change History: This Article has been retracted; see accompanying Retraction. Corrected online 20 January: In this Article, author Frank Rigo was incorrectly listed with a middle initial; this has been corrected in the online versions of the paper.

12.
Nature ; 528(7583): 517-22, 2015 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-26675721

RESUMEN

T helper 17 (TH17) lymphocytes protect mucosal barriers from infections, but also contribute to multiple chronic inflammatory diseases. Their differentiation is controlled by RORγt, a ligand-regulated nuclear receptor. Here we identify the RNA helicase DEAD-box protein 5 (DDX5) as a RORγt partner that coordinates transcription of selective TH17 genes, and is required for TH17-mediated inflammatory pathologies. Surprisingly, the ability of DDX5 to interact with RORγt and coactivate its targets depends on intrinsic RNA helicase activity and binding of a conserved nuclear long noncoding RNA (lncRNA), Rmrp, which is mutated in patients with cartilage-hair hypoplasia. A targeted Rmrp gene mutation in mice, corresponding to a gene mutation in cartilage-hair hypoplasia patients, altered lncRNA chromatin occupancy, and reduced the DDX5-RORγt interaction and RORγt target gene transcription. Elucidation of the link between Rmrp and the DDX5-RORγt complex reveals a role for RNA helicases and lncRNAs in tissue-specific transcriptional regulation, and provides new opportunities for therapeutic intervention in TH17-dependent diseases.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , ARN Largo no Codificante/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Animales , Cromatina/genética , Cromatina/metabolismo , ARN Helicasas DEAD-box/genética , Femenino , Regulación de la Expresión Génica/genética , Cabello/anomalías , Enfermedad de Hirschsprung/genética , Humanos , Síndromes de Inmunodeficiencia/genética , Inflamación/inmunología , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Especificidad de Órganos , Osteocondrodisplasias/congénito , Osteocondrodisplasias/genética , Enfermedades de Inmunodeficiencia Primaria , Unión Proteica , ARN Largo no Codificante/genética , Transcripción Genética/genética
13.
Cell Immunol ; 330: 183-187, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29807623

RESUMEN

Obesity is a worldwide public health concern yet no safe therapies are currently available. The activity of sympathetic neurons is necessary and sufficient for fat mass reduction, via norepinephrine (NE) signaling. Macrophage accumulation in the adipose tissue is thought to play the central role in the onset of obesity, yet their relation to NE has been controversial. We have identified a population of sympathetic neuron-associated macrophages (SAMs) that control obesity via the uptake and clearing of NE. Here we focus on the neuro-immune regulation of obesity by discussing the genetic, cellular and functional signatures of SAMs vis-a-vis adipose tissue macrophages (ATMs).


Asunto(s)
Tejido Adiposo/inmunología , Macrófagos/inmunología , Neuronas/inmunología , Obesidad/inmunología , Sistema Nervioso Simpático/inmunología , Adipocitos/inmunología , Tejido Adiposo/inervación , Animales , Homeostasis/inmunología , Humanos , Norepinefrina/inmunología , Norepinefrina/metabolismo , Obesidad/fisiopatología , Sistema Nervioso Simpático/fisiopatología
14.
Exp Cell Res ; 360(1): 27-30, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28342901

RESUMEN

In recent decades, obesity has become a global public health crisis irrespective of age or gender [20]. But according to historic records, concerns over appropriate maintenance of body size have been long established. For more than to 2 millennia, the main therapeutic approach to curb excess weight has been to recommend dietary restrictions and regular exercise (Haslam, 2016). Nevertheless, more contemporary studies indicate that the employment of such approaches in the treatment of severely obese patients causes metabolic adaptions which impair their long-term success in weight management [8]. These evidences highlight thus, the urgency in the search for a more comprehensive knowledge of the mechanisms that underlie the control of body weight, which would be essential for the development of effective strategies for the treatment of obesity and its comorbidities. Importantly, the discovery of the hormone leptin [33]and the use of novel techniques in targeted transgenesis [32] have enabled progress in defining some of the key players and the molecular mechanisms that are involved in the processes that control body size homeostasis and energy balance, and how obesity may disrupt leptin's feedback loop and lead to the pathology of metabolic syndrome. On the light of such findings, here we review how the sympathetic nervous system modulates adipose tissue metabolism downstream of leptin's action on the CNS, with particular focus on how this system may be disrupted in the context of excess adiposity, plus highlight the potential clinical implications arising from a better understanding of the physiologic control of the sympathetic neuro-adipose connection.


Asunto(s)
Tejido Adiposo/fisiología , Peso Corporal , Metabolismo Energético , Sistema Nervioso Simpático/fisiología , Animales , Humanos
16.
Am J Physiol Endocrinol Metab ; 323(4): E333-E335, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36044709
17.
J Immunol ; 194(11): 5253-60, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25917102

RESUMEN

The hormone leptin plays a key role in energy homeostasis, and the absence of either leptin or its receptor (LepR) leads to severe obesity and metabolic disorders. To avoid indirect effects and to address the cell-intrinsic role of leptin signaling in the immune system, we conditionally targeted LepR in T cells. In contrast with pleiotropic immune disorders reported in obese mice with leptin or LepR deficiency, we found that LepR deficiency in CD4(+) T cells resulted in a selective defect in both autoimmune and protective Th17 responses. Reduced capacity for differentiation toward a Th17 phenotype by lepr-deficient T cells was attributed to reduced activation of the STAT3 and its downstream targets. This study establishes cell-intrinsic roles for LepR signaling in the immune system and suggests that leptin signaling during T cell differentiation plays a crucial role in T cell peripheral effector function.


Asunto(s)
Diferenciación Celular/inmunología , Leptina/inmunología , Obesidad/inmunología , Receptores de Leptina/inmunología , Células Th17/citología , Animales , Autoinmunidad/genética , Autoinmunidad/inmunología , Diferenciación Celular/genética , Células Cultivadas , Citrobacter rodentium/inmunología , Colitis/inmunología , Infecciones por Enterobacteriaceae/inmunología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Receptores de Leptina/genética , Factor de Transcripción STAT3/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Células Th17/inmunología
19.
EMBO Rep ; 15(10): 1023-35, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25239948

RESUMEN

People think they are in control of their own decisions: what to eat or drink, whom to marry or pick a fight with, where to live, what to buy. Behavioural economists and neurophysiologists have long studied decision-making behaviours. However, these behaviours have only recently been studied through the light of molecular genetics. Here, we review recent research in mice, Drosophila melanogaster and Caenorhabditis elegans, that analyses the molecular and cellular mechanisms underlying decision-making. These studies interrogate decision-making about food, sexual behaviour, aggression or foraging strategies, and add molecular and cell biology understanding onto the consilience of brain and decision.


Asunto(s)
Caenorhabditis elegans/genética , Toma de Decisiones/fisiología , Drosophila melanogaster/genética , Animales , Encéfalo/metabolismo , Encéfalo/fisiología , Caenorhabditis elegans/fisiología , Drosophila melanogaster/fisiología , Conducta Alimentaria/fisiología , Humanos , Ratones , Conducta Sexual Animal/fisiología
20.
bioRxiv ; 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38798460

RESUMEN

T cells have emerged as sex-dependent orchestrators of pain chronification but the sexually dimorphic mechanisms by which T cells control pain sensitivity is not resolved. Here, we demonstrate an influence of regulatory T cells (Tregs) on pain processing that is distinct from their canonical functions of immune regulation and tissue repair. Specifically, meningeal Tregs (mTregs) express the endogenous opioid, enkephalin, and mTreg-derived enkephalin exerts an antinociceptive action through a presynaptic opioid receptor signaling mechanism that is dispensable for immunosuppression. mTregs are both necessary and sufficient for suppressing mechanical pain sensitivity in female but not male mice. Notably, the mTreg modulation of pain thresholds depends on sex-hormones and expansion of enkephalinergic mTregs during gestation imparts a remarkable pregnancy-induced analgesia in a pre-existing, chronic, unremitting neuropathic pain model. These results uncover a fundamental sex-specific, pregnancy-pronounced, and immunologically-derived endogenous opioid circuit for nociceptive regulation with critical implications for pain biology and maternal health.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA