Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Metab ; 34(8): 1183-1200.e12, 2022 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-35841892

RESUMO

Hypoglycemia is a clinical hallmark of severe malaria, the often-lethal outcome of Plasmodium falciparum infection. Here, we report that malaria-associated hypoglycemia emerges from a non-canonical resistance mechanism, whereby the infected host reduces glycemia to starve Plasmodium. This hypometabolic response is elicited by labile heme, a byproduct of hemolysis that induces illness-induced anorexia and represses hepatic glucose production. While transient repression of hepatic glucose production prevents unfettered immune-mediated inflammation, organ damage, and anemia, when sustained over time it leads to hypoglycemia, compromising host energy expenditure and adaptive thermoregulation. The latter arrests the development of asexual stages of Plasmodium via a mechanism associated with parasite mitochondrial dysfunction. In response, Plasmodium activates a transcriptional program associated with the reduction of virulence and sexual differentiation toward the generation of transmissible gametocytes. In conclusion, malaria-associated hypoglycemia represents a trade-off of a hypometabolic-based defense strategy that balances parasite virulence versus transmission.


Assuntos
Hipoglicemia , Malária Falciparum , Malária , Glucose , Humanos , Malária Falciparum/parasitologia , Plasmodium falciparum
2.
Nature ; 597(7876): 410-414, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34408322

RESUMO

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.


Assuntos
Tecido Adiposo/inervação , Tecido Adiposo/metabolismo , Encéfalo/metabolismo , Imunidade Inata/imunologia , Mesoderma/citologia , Vias Neurais , Neurônios/citologia , Obesidade/metabolismo , Tecido Adiposo/citologia , Animais , Encéfalo/citologia , Sinais (Psicologia) , Citocinas/metabolismo , Metabolismo Energético , Feminino , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Gônadas/metabolismo , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Sistema Nervoso Simpático/citologia , Sistema Nervoso Simpático/metabolismo
3.
FEBS J ; 287(21): 4602-4606, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32500671

RESUMO

The 'Crosstalks of immunity and metabolism' Symposium was focused on how the intercommunication between different organs and the immune system affects organismal health. At this meeting, experts in immunology and metabolic research provided novel insights into the growing field of immunometabolism. This report attempts to review and integrate views, ideas, propositions, and conclusions that emanated from the symposium.


Assuntos
Metabolismo Energético/imunologia , Sistema Imunitário/imunologia , Imunidade Inata/imunologia , Inflamação/imunologia , Animais , Humanos
4.
Cell Metab ; 31(6): 1120-1135.e7, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32402266

RESUMO

Anti-obesity drugs in the amphetamine (AMPH) class act in the brain to reduce appetite and increase locomotion. They are also characterized by adverse cardiovascular effects with origin that, despite absence of any in vivo evidence, is attributed to a direct sympathomimetic action in the heart. Here, we show that the cardiac side effects of AMPH originate from the brain and can be circumvented by PEGylation (PEGyAMPH) to exclude its central action. PEGyAMPH does not enter the brain and facilitates SNS activity via theß2-adrenoceptor, protecting mice against obesity by increasing lipolysis and thermogenesis, coupled to higher heat dissipation, which acts as an energy sink to increase energy expenditure without altering food intake or locomotor activity. Thus, we provide proof-of-principle for a novel class of exclusively peripheral anti-obesity sympathofacilitators that are devoid of any cardiovascular and brain-related side effects.


Assuntos
Anfetamina/farmacologia , Fármacos Antiobesidade/farmacologia , Encéfalo/efeitos dos fármacos , Obesidade/tratamento farmacológico , Animais , Encéfalo/metabolismo , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Obesidade/metabolismo
5.
Mol Metab ; 24: 64-79, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30954544

RESUMO

OBJECTIVE: The ferritin heavy/heart chain (FTH) gene encodes the ferroxidase component of the iron (Fe) sequestering ferritin complex, which plays a central role in the regulation of cellular Fe metabolism. Here we tested the hypothesis that ferritin regulates organismal Fe metabolism in a manner that impacts energy balance and thermal homeostasis. METHODS: We developed a mouse strain, referred herein as FthR26 fl/fl, expressing a tamoxifen-inducible Cre recombinase under the control of the Rosa26 (R26) promoter and carrying two LoxP (fl) sites: one at the 5'end of the Fth promoter and another the 3' end of the first Fth exon. Tamoxifen administration induces global deletion of Fth in adult FthR26Δ/Δ mice, testing whether FTH is required for maintenance of organismal homeostasis. RESULTS: Under standard nutritional Fe supply, Fth deletion in adult FthR26Δ/Δ mice led to a profound deregulation of organismal Fe metabolism, oxidative stress, inflammation, and multi-organ damage, culminating in death. Unexpectedly, Fth deletion was also associated with a profound atrophy of white and brown adipose tissue as well as with collapse of energy expenditure and thermogenesis. This was attributed mechanistically to mitochondrial dysfunction, as assessed in the liver and in adipose tissue. CONCLUSION: The FTH component of ferritin acts as a master regulator of organismal Fe homeostasis, coupling nutritional Fe supply to organismal redox homeostasis, energy expenditure and thermoregulation.


Assuntos
Metabolismo Energético , Ferritinas/metabolismo , Termogênese , Tecido Adiposo/metabolismo , Animais , Células Cultivadas , Ferritinas/genética , Deleção de Genes , Hepatócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Estresse Oxidativo
6.
Nat Med ; 23(11): 1309-1318, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29035364

RESUMO

The cellular mechanism(s) linking macrophages to norepinephrine (NE)-mediated regulation of thermogenesis have been a topic of debate. Here we identify sympathetic neuron-associated macrophages (SAMs) as a population of cells that mediate clearance of NE via expression of solute carrier family 6 member 2 (SLC6A2), an NE transporter, and monoamine oxidase A (MAOA), a degradation enzyme. Optogenetic activation of the sympathetic nervous system (SNS) upregulates NE uptake by SAMs and shifts the SAM profile to a more proinflammatory state. NE uptake by SAMs is prevented by genetic deletion of Slc6a2 or inhibition of the encoded transporter. We also observed an increased proportion of SAMs in the SNS of two mouse models of obesity. Genetic ablation of Slc6a2 in SAMs increases brown adipose tissue (BAT) content, causes browning of white fat, increases thermogenesis, and leads to substantial and sustained weight loss in obese mice. We further show that this pathway is conserved, as human sympathetic ganglia also contain SAMs expressing the analogous molecular machinery for NE clearance, which thus constitutes a potential target for obesity treatment.


Assuntos
Macrófagos/metabolismo , Neurônios/metabolismo , Norepinefrina/metabolismo , Obesidade/patologia , Sistema Nervoso Simpático/patologia , Animais , Receptor 1 de Quimiocina CX3C/metabolismo , Perfilação da Expressão Gênica , Homeostase , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/genética , Obesidade/genética
8.
Nat Commun ; 8: 14967, 2017 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-28367972

RESUMO

Conditional expression of diphtheria toxin receptor (DTR) is widely used for tissue-specific ablation of cells. However, diphtheria toxin (DT) crosses the blood-brain barrier, which limits its utility for ablating peripheral cells using Cre drivers that are also expressed in the central nervous system (CNS). Here we report the development of a brain-sparing DT, termed BRAINSPAReDT, for tissue-specific genetic ablation of cells outside the CNS. We prevent blood-brain barrier passage of DT through PEGylation, which polarizes the molecule and increases its size. We validate BRAINSPAReDT with regional genetic sympathectomy: BRAINSPAReDT ablates peripheral but not central catecholaminergic neurons, thus avoiding the Parkinson-like phenotype associated with full dopaminergic depletion. Regional sympathectomy compromises adipose tissue thermogenesis, and renders mice susceptible to obesity. We provide a proof of principle that BRAINSPAReDT can be used for Cre/DTR tissue-specific ablation outside the brain using CNS drivers, while consolidating the link between adiposity and the sympathetic nervous system.


Assuntos
Encéfalo/citologia , Linhagem da Célula , Toxina Diftérica/metabolismo , Deleção de Genes , Animais , Toxina Diftérica/química , Suscetibilidade a Doenças , Metabolismo Energético , Comportamento Alimentar , Feminino , Regulação da Expressão Gênica , Intolerância à Glucose/complicações , Intolerância à Glucose/patologia , Células HeLa , Humanos , Hidrodinâmica , Masculino , Camundongos , Movimento , Neurônios/metabolismo , Norepinefrina/metabolismo , Obesidade/complicações , Obesidade/patologia , Doença de Parkinson/patologia , Doença de Parkinson/fisiopatologia , Polietilenoglicóis/química , Simpatectomia , Termogênese/genética
10.
Exp Cell Res ; 360(1): 27-30, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28342901

RESUMO

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.


Assuntos
Tecido Adiposo/fisiologia , Peso Corporal , Metabolismo Energético , Sistema Nervoso Simpático/fisiologia , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...