Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Brain Behav Immun ; 117: 330-346, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38309640

RESUMO

Nutrient composition in obesogenic diets may influence the severity of disorders associated with obesity such as insulin-resistance and chronic inflammation. Here we hypothesized that obesogenic diets rich in fat and varying in fatty acid composition, particularly in omega 6 (ω6) to omega 3 (ω3) ratio, have various effects on energy metabolism, neuroinflammation and behavior. Mice were fed either a control diet or a high fat diet (HFD) containing either low (LO), medium (ME) or high (HI) ω6/ω3 ratio. Mice from the HFD-LO group consumed less calories and exhibited less body weight gain compared to other HFD groups. Both HFD-ME and HFD-HI impaired glucose metabolism while HFD-LO partly prevented insulin intolerance and was associated with normal leptin levels despite higher subcutaneous and perigonadal adiposity. Only HFD-HI increased anxiety and impaired spatial memory, together with increased inflammation in the hypothalamus and hippocampus. Our results show that impaired glucose metabolism and neuroinflammation are uncoupled, and support that diets with a high ω6/ω3 ratio are associated with neuroinflammation and the behavioral deterioration coupled with the consumption of diets rich in fat.


Assuntos
Insulinas , Doenças Neuroinflamatórias , Animais , Camundongos , Obesidade/metabolismo , Dieta Hiperlipídica/efeitos adversos , Ácidos Graxos/metabolismo , Inflamação , Glucose
2.
Glia ; 69(1): 42-60, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32659044

RESUMO

In humans, obesity is associated with brain inflammation, glial reactivity, and immune cells infiltration. Studies in rodents have shown that glial reactivity occurs within 24 hr of high-fat diet (HFD) consumption, long before obesity development, and takes place mainly in the hypothalamus (HT), a crucial brain structure for controlling body weight. Here, we sought to characterize the postprandial HT inflammatory response to 1, 3, and 6 hr of exposure to either a standard diet (SD) or HFD. HFD exposure increased gene expression of astrocyte and microglial markers (glial fibrillary acidic protein [GFAP] and Iba1, respectively) compared to SD-treated mice and induced morphological modifications of microglial cells in HT. This remodeling was associated with higher expression of inflammatory genes and differential regulation of hypothalamic neuropeptides involved in energy balance regulation. DREADD and PLX5622 technologies, used to modulate GFAP-positive or microglial cells activity, respectively, showed that both glial cell types are involved in hypothalamic postprandial inflammation, with their own specific kinetics and reactiveness to ingested foods. Thus, recurrent exacerbated postprandial inflammation in the brain might promote obesity and needs to be characterized to address this worldwide crisis.


Assuntos
Gorduras na Dieta , Microglia , Animais , Dieta Hiperlipídica/efeitos adversos , Proteína Glial Fibrilar Ácida , Hipotálamo , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade
3.
J Neurosci Res ; 98(10): 2045-2071, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32530066

RESUMO

Melanin-concentrating hormone (MCH) is a ubiquitous vertebrate neuropeptide predominantly synthesized by neurons of the diencephalon that can act through two G protein-coupled receptors, called MCHR1 and MCHR2. The expression of Mchr1 has been investigated in both rats and mice, but its synthesis remains poorly described. After identifying an antibody that detects MCHR1 with high specificity, we employed immunohistochemistry to map the distribution of MCHR1 in the CNS of rats and mice. Multiple neurochemical markers were also employed to characterize some of the neuronal populations that synthesize MCHR1. Our results show that MCHR1 is abundantly found in a subcellular structure called the primary cilium, which has been associated, among other functions, with the detection of free neurochemical messengers present in the extracellular space. Ciliary MCHR1 was found in a wide range of areas, including the olfactory bulb, cortical mantle, striatum, hippocampal formation, amygdala, midline thalamic nuclei, periventricular hypothalamic nuclei, midbrain areas, and in the spinal cord. No differences were observed between male and female mice, and interspecies differences were found in the caudate-putamen nucleus and the subgranular zone. Ciliary MCHR1 was found in close association with several neurochemical markers, including tyrosine hydroxylase, calretinin, kisspeptin, estrogen receptor, oxytocin, vasopressin, and corticotropin-releasing factor. Given the role of neuronal primary cilia in sensing free neurochemical messengers in the extracellular fluid, the widespread distribution of ciliary MCHR1, and the diverse neurochemical populations who synthesize MCHR1, our data indicate that nonsynaptic communication plays a prominent role in the normal function of the MCH system.


Assuntos
Encéfalo/metabolismo , Cílios/metabolismo , Receptores de Somatostatina/biossíntese , Caracteres Sexuais , Animais , Cílios/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Receptores de Somatostatina/genética
4.
EMBO Rep ; 17(12): 1738-1752, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27733491

RESUMO

Sickness behavior defines the endocrine, autonomic, behavioral, and metabolic responses associated with infection. While inflammatory responses were suggested to be instrumental in the loss of appetite and body weight, the molecular underpinning remains unknown. Here, we show that systemic or central lipopolysaccharide (LPS) injection results in specific hypothalamic changes characterized by a precocious increase in the chemokine ligand 2 (CCL2) followed by an increase in pro-inflammatory cytokines and a decrease in the orexigenic neuropeptide melanin-concentrating hormone (MCH). We therefore hypothesized that CCL2 could be the central relay for the loss in body weight induced by the inflammatory signal LPS. We find that central delivery of CCL2 promotes neuroinflammation and the decrease in MCH and body weight. MCH neurons express CCL2 receptor and respond to CCL2 by decreasing both electrical activity and MCH release. Pharmacological or genetic inhibition of CCL2 signaling opposes the response to LPS at both molecular and physiologic levels. We conclude that CCL2 signaling onto MCH neurons represents a core mechanism that relays peripheral inflammation to sickness behavior.


Assuntos
Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Hormônios Hipotalâmicos/metabolismo , Hipotálamo/metabolismo , Inflamação/metabolismo , Melaninas/metabolismo , Neurônios/metabolismo , Hormônios Hipofisários/metabolismo , Transdução de Sinais , Animais , Quimiocina CCL2/deficiência , Quimiocina CCL2/imunologia , Citocinas/biossíntese , Citocinas/genética , Citocinas/imunologia , Hormônios Hipotalâmicos/genética , Hormônios Hipotalâmicos/imunologia , Comportamento de Doença , Lipopolissacarídeos/imunologia , Melaninas/genética , Melaninas/imunologia , Camundongos , Neurônios/imunologia , Hormônios Hipofisários/genética , Hormônios Hipofisários/imunologia , Receptores CCR2/metabolismo , Redução de Peso
5.
J Neurosci ; 33(28): 11643-54, 2013 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-23843532

RESUMO

CXCR4, a receptor for the chemokine CXCL12 (stromal-cell derived factor-1α), is a G-protein-coupled receptor (GPCR), expressed in the immune and CNS and integrally involved in various neurological disorders. The GABAB receptor is also a GPCR that mediates metabotropic action of the inhibitory neurotransmitter GABA and is located on neurons and immune cells as well. Using diverse approaches, we report novel interaction between GABAB receptor agents and CXCR4 and demonstrate allosteric binding of these agents to CXCR4. First, both GABAB antagonists and agonists block CXCL12-elicited chemotaxis in human breast cancer cells. Second, a GABAB antagonist blocks the potentiation by CXCL12 of high-threshold Ca(2+) channels in rat neurons. Third, electrophysiology in Xenopus oocytes and human embryonic kidney cell line 293 cells in which we coexpressed rat CXCR4 and the G-protein inward rectifier K(+) (GIRK) channel showed that GABAB antagonist and agonist modified CXCL12-evoked activation of GIRK channels. To investigate whether GABAB ligands bind to CXCR4, we expressed this receptor in heterologous systems lacking GABAB receptors and performed competition binding experiments. Our fluorescent resonance energy transfer experiments suggest that GABAB ligands do not bind CXCR4 at the CXCL12 binding pocket suggesting allosteric modulation, in accordance with our electrophysiology experiments. Finally, using backscattering interferometry and lipoparticles containing only the CXCR4 receptor, we quantified the binding affinity for the GABAB ligands, confirming a direct interaction with the CXCR4 receptor. The effect of GABAergic agents on CXCR4 suggests new therapeutic potentials for neurological and immune diseases.


Assuntos
Baclofeno/farmacologia , Quimiocina CXCL12/metabolismo , Agonistas dos Receptores de GABA-B/metabolismo , Receptores CXCR4/metabolismo , Regulação Alostérica/efeitos dos fármacos , Regulação Alostérica/fisiologia , Animais , Baclofeno/metabolismo , Linhagem Celular Tumoral , Feminino , GABAérgicos/metabolismo , Agonistas dos Receptores de GABA-B/farmacologia , Células HEK293 , Humanos , Masculino , Técnicas de Cultura de Órgãos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Ratos , Ratos Wistar , Xenopus laevis
6.
PLoS Biol ; 8(4): e1000355, 2010 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-20405001

RESUMO

Current antidepressant treatments are inadequate for many individuals, and when they are effective, they require several weeks of administration before a therapeutic effect can be observed. Improving the treatment of depression is challenging. Recently, the two-pore domain potassium channel TREK-1 has been identified as a new target in depression, and its antagonists might become effective antidepressants. In mice, deletion of the TREK-1 gene results in a depression-resistant phenotype that mimics antidepressant treatments. Here, we validate in mice the antidepressant effects of spadin, a secreted peptide derived from the propeptide generated by the maturation of the neurotensin receptor 3 (NTSR3/Sortilin) and acting through TREK-1 inhibition. NTSR3/Sortilin interacted with the TREK-1 channel, as shown by immunoprecipitation of TREK-1 and NTSR3/Sortilin from COS-7 cells and cortical neurons co-expressing both proteins. TREK-1 and NTSR3/Sortilin were colocalized in mouse cortical neurons. Spadin bound specifically to TREK-1 with an affinity of 10 nM. Electrophysiological studies showed that spadin efficiently blocked the TREK-1 activity in COS-7 cells, cultured hippocampal pyramidal neurons, and CA3 hippocampal neurons in brain slices. Spadin also induced in vivo an increase of the 5-HT neuron firing rate in the Dorsal Raphe Nucleus. In five behavioral tests predicting an antidepressant response, spadin-treated mice showed a resistance to depression as found in TREK-1 deficient mice. More importantly, an intravenous 4-d treatment with spadin not only induced a strong antidepressant effect but also enhanced hippocampal phosphorylation of CREB protein and neurogenesis, considered to be key markers of antidepressant action after chronic treatment with selective serotonin reuptake inhibitors. This work also shows the development of a reliable method for dosing the propeptide in serum of mice by using AlphaScreen technology. These findings point out spadin as a putative antidepressant of new generation with a rapid onset of action. Spadin can be regarded as the first natural antidepressant peptide identified. It corresponds to a new concept to address the treatment of depression.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Antidepressivos/química , Peptídeos/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/química , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/farmacologia , Animais , Antidepressivos/metabolismo , Antidepressivos/uso terapêutico , Células COS , Chlorocebus aethiops , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Transtorno Depressivo/tratamento farmacológico , Desenho de Fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Técnicas de Patch-Clamp , Peptídeos/química , Peptídeos/genética , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Bloqueadores dos Canais de Potássio/metabolismo , Canais de Potássio de Domínios Poros em Tandem/antagonistas & inibidores , Canais de Potássio de Domínios Poros em Tandem/genética , Núcleos da Rafe/efeitos dos fármacos , Serotonina/metabolismo , Transmissão Sináptica/efeitos dos fármacos
7.
Antioxid Redox Signal ; 37(4-6): 349-369, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35166124

RESUMO

Aims: Although prebiotics, probiotics, and fecal transplantation can alter the sensation of hunger and/or feeding behavior, the role of the constitutive gut microbiota in the short-term regulation of food intake during normal physiology is still unclear. Results: An antibiotic-induced microbiota depletion study was designed to compare feeding behavior in conventional and microbiota-depleted mice. Tissues were sampled to characterize the time profile of microbiota-derived signals in mice during consumption of either standard or high-fat food for 1 h. Pharmacological and genetic tools were used to evaluate the contribution of postprandial endotoxemia and inflammatory responses in the short-term regulation of food intake. We observed constitutive microbial and macronutrient-dependent control of food intake at the time scale of a meal; that is, within 1 h of food introduction. Specifically, microbiota depletion increased food intake, and the microbiota-derived anorectic effect became significant during the consumption of high-fat but not standard food. This anorectic effect correlated with a specific postprandial microbial metabolic signature, and did not require postprandial endotoxemia or an NOD-, LRR-, and Pyrin domain-containing protein 3-inflammasome-mediated inflammatory response. Innovation and Conclusion: These findings show that the gut microbiota controls host appetite at the time scale of a meal under normal physiology. Interestingly, a microbiota-derived anorectic effect develops specifically with a high-fat meal, indicating that gut microbiota activity is involved in the satietogenic properties of foods. Antioxid. Redox Signal. 37, 349-369.


Assuntos
Depressores do Apetite , Endotoxemia , Microbiota , Animais , Ingestão de Alimentos , Peptídeo 1 Semelhante ao Glucagon , Inflamação , Camundongos , Camundongos Endogâmicos NOD , Estresse Oxidativo
8.
PLoS One ; 16(4): e0243333, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33852580

RESUMO

The emergence and quick spread of SARS-CoV-2 has pointed at a low capacity response for testing large populations in many countries, in line of material, technical and staff limitations. The traditional RT-qPCR diagnostic test remains the reference method and is by far the most widely used test. These assays are limited to a few probe sets, require large sample PCR reaction volumes, along with an expensive and time-consuming RNA extraction step. Here we describe a quantitative nanofluidic assay that overcomes some of these shortcomings, based on the BiomarkTM instrument from Fluidigm. This system offers the possibility of performing 4608 qPCR end-points in a single run, equivalent to 192 clinical samples combined with 12 pairs of primers/probe sets in duplicate, thus allowing the monitoring of SARS-CoV-2 including the detection of specific SARS-CoV-2 variants, as well as the detection other pathogens and/or host cellular responses (virus receptors, response markers, microRNAs). The 10 nL-range volume of BiomarkTM reactions is compatible with sensitive and reproducible reactions that can be easily and cost-effectively adapted to various RT-qPCR configurations and sets of primers/probe. Finally, we also evaluated the use of inactivating lysis buffers composed of various detergents in the presence or absence of proteinase K to assess the compatibility of these buffers with a direct reverse transcription enzymatic step and we propose several protocols, bypassing the need for RNA purification. We advocate that the combined utilization of an optimized processing buffer and a high-throughput real-time PCR device would contribute to improve the turn-around-time to deliver the test results to patients and increase the SARS-CoV-2 testing capacities.


Assuntos
COVID-19/diagnóstico , Técnicas Analíticas Microfluídicas/métodos , SARS-CoV-2/isolamento & purificação , Manejo de Espécimes/métodos , Adulto , COVID-19/virologia , Teste para COVID-19/métodos , Primers do DNA , Testes Diagnósticos de Rotina/métodos , Feminino , Humanos , Masculino , MicroRNAs/genética , RNA Viral/genética , Reação em Cadeia da Polimerase em Tempo Real/métodos , SARS-CoV-2/genética , Sensibilidade e Especificidade
9.
J Neurosci ; 29(8): 2528-33, 2009 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-19244527

RESUMO

Glucose sensing by hypothalamic neurons triggers adaptive metabolic and behavioral responses. In orexin neurons, extracellular glucose activates a leak K(+) current promoting electrical activity inhibition. Sensitivity to external acidification and halothane, and resistance to ruthenium red designated the tandem-pore K(+) (K(2P)) channel subunit TASK3 as part of the glucose-induced channel. Here, we show that glucose inhibition and its pH sensitivity persist in mice lacking TASK3 or TASK1, or both subunits. We also tested the implication of another class of K(2P) channels activated by halothane. In the corresponding TREK1/2/TRAAK triple knock-out mice, glucose inhibition persisted in hypothalamic neurons ruling out a major contribution of these subunits to the glucose-activated K(+) conductance. Finally, block of this glucose-induced hyperpolarizing current by low Ba(2+) concentrations was consistent with the conclusion that K(2P) channels are not required for glucosensing in hypothalamic neurons.


Assuntos
Glucose/farmacologia , Hipotálamo/citologia , Inibição Neural/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Canais de Potássio de Domínios Poros em Tandem/deficiência , Edulcorantes/farmacologia , Animais , Bário/farmacologia , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Inibição Neural/fisiologia , Neurônios/fisiologia , Neuropeptídeos/metabolismo , Orexinas , Técnicas de Patch-Clamp/métodos , Canais de Potássio/deficiência , Canais de Potássio de Domínios Poros em Tandem/classificação
10.
Neurobiol Dis ; 38(3): 386-94, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20206263

RESUMO

The presymptomatic phase of Parkinson's disease (PD) is now recognized as a prodromal phase, with compensatory mechanism masking its progression and non-motor early manifestations, such as depression, cognitive disturbances and apathy. Those mechanisms were thought to be strictly dopamine-mediated until recent advances have shed light upon involvement of putative outside-basal ganglia, i.e. cortical, structures. We took advantage of our progressive 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated macaque model to monitor whole genome transcriptional changes in several brain areas. Our data reveals that transcriptomic activity changes take place from early stages, suggesting very early compensatory mechanisms or pathological activity outside the basal ganglia, including the PFC. Specific transcriptomic changes occurring in the PFC of fully parkinsonian MPTP-treated macaques have been identified. Interestingly, a large part of these transcriptomic changes were also observed in human post-mortem samples of patients with neurodegenerative diseases analysed by quantitative PCR. These results suggest that the PFC is able to detect the progression of dopamine denervation even at very early time points. There are therefore mechanisms, within the PFC, leading to compensatory alterations and/or participating to pathophysiology of prodromal PD manifestations.


Assuntos
Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Transtornos Parkinsonianos/genética , Transtornos Parkinsonianos/metabolismo , Córtex Pré-Frontal/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Globo Pálido/metabolismo , Humanos , Macaca fascicularis , Masculino , Pessoa de Meia-Idade , Modelos Neurológicos , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Putamen/metabolismo , RNA Mensageiro/metabolismo , Especificidade da Espécie , Tálamo/metabolismo
11.
Cell Rep ; 30(9): 3067-3078.e5, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32130907

RESUMO

Mechanistic studies in rodents evidenced synaptic remodeling in neuronal circuits that control food intake. However, the physiological relevance of this process is not well defined. Here, we show that the firing activity of anorexigenic POMC neurons located in the hypothalamus is increased after a standard meal. Postprandial hyperactivity of POMC neurons relies on synaptic plasticity that engages pre-synaptic mechanisms, which does not involve structural remodeling of synapses but retraction of glial coverage. These functional and morphological neuroglial changes are triggered by postprandial hyperglycemia. Chemogenetically induced glial retraction on POMC neurons is sufficient to increase POMC activity and modify meal patterns. These findings indicate that synaptic plasticity within the melanocortin system happens at the timescale of meals and likely contributes to short-term control of food intake. Interestingly, these effects are lost with a high-fat meal, suggesting that neuroglial plasticity of POMC neurons is involved in the satietogenic properties of foods.


Assuntos
Hiperglicemia/fisiopatologia , Hipotálamo/metabolismo , Refeições , Neuroglia/patologia , Plasticidade Neuronal , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Animais , Glicemia/metabolismo , Fenômenos Eletrofisiológicos , Comportamento Alimentar , Hiperglicemia/sangue , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Período Pós-Prandial , Sinapses/metabolismo
12.
BMC Evol Biol ; 8: 330, 2008 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-19068116

RESUMO

BACKGROUND: Brain-expressed genes that were created in primate lineage represent obvious candidates to investigate molecular mechanisms that contributed to neural reorganization and emergence of new behavioural functions in Homo sapiens. PMCHL1 arose from retroposition of a pro-melanin-concentrating hormone (PMCH) antisense mRNA on the ancestral human chromosome 5p14 when platyrrhines and catarrhines diverged. Mutations before divergence of hylobatidae led to creation of new exons and finally PMCHL1 duplicated in an ancestor of hominids to generate PMCHL2 at the human chromosome 5q13. A complex pattern of spliced and unspliced PMCHL RNAs were found in human brain and testis. RESULTS: Several novel spliced PMCHL transcripts have been characterized in human testis and fetal brain, identifying an additional exon and novel splice sites. Sequencing of PMCHL genes in several non-human primates allowed to carry out phylogenetic analyses revealing that the initial retroposition event took place within an intron of the brain cadherin (CDH12) gene, soon after platyrrhine/catarrhine divergence, i.e. 30-35 Mya, and was concomitant with the insertion of an AluSg element. Sequence analysis of the spliced PMCHL transcripts identified only short ORFs of less than 300 bp, with low (VMCH-p8 and protein variants) or no evolutionary conservation. Western blot analyses of human and macaque tissues expressing PMCHL RNA failed to reveal any protein corresponding to VMCH-p8 and protein variants encoded by spliced transcripts. CONCLUSION: Our present results improve our knowledge of the gene structure and the evolutionary history of the primate-specific chimeric PMCHL genes. These genes produce multiple spliced transcripts, bearing short, non-conserved and apparently non-translated ORFs that may function as mRNA-like non-coding RNAs.


Assuntos
Hormônios Hipotalâmicos/genética , Precursores de Proteínas/genética , Splicing de RNA , RNA não Traduzido/genética , Adulto , Sequência de Aminoácidos , Animais , Encéfalo/metabolismo , Cromossomos Humanos Par 5/genética , Evolução Molecular , Éxons , Humanos , Íntrons , Macaca/genética , Masculino , Dados de Sequência Molecular , Filogenia , Primatas , RNA não Traduzido/metabolismo , Alinhamento de Sequência , Testículo/metabolismo
13.
J Neuroimmunol ; 198(1-2): 46-55, 2008 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-18547650

RESUMO

Following inflammation or infection, cytokines are released in the blood. Besides their effect on the immune system, cytokines can also act in the brain to modulate our behaviors, inducing for example anorexia when produced in large amount. This review focuses on our current knowledge on how cytokines can influence the brain and the behaviors through several possible pathways: modulating peripheral neurons which project to the brain through the vagus nerve, modulating the levels of hormones such as leptin which can act to the brain through the humoral pathway and possibly acting directly in the brain, through the local production of cytokines and chemokines such as SDF-1alpha/CXCL12.


Assuntos
Encéfalo/fisiologia , Quimiocinas/fisiologia , Citocinas/fisiologia , Vias Aferentes/fisiologia , Animais , Encéfalo/citologia , Hormônios/metabolismo , Humanos , Modelos Biológicos , Neurônios Aferentes/fisiologia
15.
Brain Struct Funct ; 223(8): 3739-3755, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30054744

RESUMO

The orexin-immunoreactive neurons are part of an important arousal-promoting hypothalamic population. Several groups have investigated these neurons during the lactation period, when numerous physiological alterations occur in the dam's body to cope with the newly acquired metabolic needs of the litter. Although those studies have probed this population during the early and intermediate stages of lactation, few works have examined its response to weaning, including the cessation of the tactile suckling stimulus as the litter stops nursing. Using double immunohistochemistry for orexin and FOS combined with three-dimensional reconstruction techniques, we investigated orexin-synthesizing neurons and their activation at different times during weaning, in addition to the role played by the suckling stimulus. We report here that weaning promoted a decline in the anterior population of orexin-immunoreactive neurons and decreased the number of double orexin-FOS neurons labeled in the central dorsomedial hypothalamus, in addition to reducing the overall number of FOS-immunoreactive cells in the whole tuberal hypothalamus. Disruption of the suckling stimulus from the pups impaired the decrease in the number of anteriorly located orexin-immunoreactive neurons, attenuated the activation of orexin-synthesizing cells in the dorsomedial hypothalamus and reduced the number of FOS-immunoreactive neurons across the tuberal hypothalamus. When taken together, our data suggest that the weaning period is necessary to restore neurochemical pathways altered during the lactation period and that the suckling stimulus plays a significant role in this process.


Assuntos
Hipotálamo/crescimento & desenvolvimento , Lactação , Neurônios/metabolismo , Orexinas/metabolismo , Desmame , Animais , Animais Lactentes , Contagem de Células , Feminino , Hipotálamo/metabolismo , Masculino , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos Wistar
16.
Regul Pept ; 143(1-3): 104-8, 2007 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-17537530

RESUMO

The mRNA encoding prepro-Melanin concentrating hormone (ppMCH) is mainly expressed in the central nervous system but has also been detected at lower amount in many peripheral tissues including spleen and thymus. At the peptide level however, several forms of the precursor can be detected in these tissues and are sometimes expressed at similar levels compared to brain. In the present work, we have studied the in vitro action of a wide range of concentration (1 nM to 1 microM) of the different peptides encoded by ppMCH i.e. neuropeptide glycine-glutamic acid (NGE), neuropeptide glutamic acid-isoleucine (NEI), Melanin concentrating hormone (MCH) and the dipeptide NEI-MCH on peripheral blood mononuclear cells (PBMC) proliferation and cytokine production following anti-CD3 stimulation. Among them only MCH decreased PBMC proliferation with a maximal effect of 35% at 100 nM. Moreover as demonstrated by using ELISA, MCH significantly decreases IL-2 production by 25% but not IL-4, INF-gamma or TNF-alpha expression. Interestingly, exogenous IL-2 decreases significantly MCH-mediated inhibition, suggesting that it is an important downstream mediator of MCH action. Finally, we showed that after 7 to 9 days of incubation, MCH also inhibits proliferation of non-stimulated PBMC. Altogether, these data demonstrate that fully mature MCH modulates proliferation of anti-CD3 stimulated PBMC partially through regulation of IL-2 production.


Assuntos
Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Hormônios Hipotalâmicos/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Melaninas/farmacologia , Hormônios Hipofisários/farmacologia , Células Cultivadas , Humanos , Interferon gama/metabolismo , Interleucina-2/metabolismo , Interleucina-2/farmacologia , Interleucina-4/metabolismo , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Fragmentos de Peptídeos/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
17.
Artigo em Inglês | MEDLINE | ID: mdl-28855891

RESUMO

The hypothalamus is a key brain region in the regulation of energy balance as it controls food intake and both energy storage and expenditure through integration of humoral, neural, and nutrient-related signals and cues. Many years of research have focused on the regulation of energy balance by hypothalamic neurons, but the most recent findings suggest that neurons and glial cells, such as microglia and astrocytes, in the hypothalamus actually orchestrate together several metabolic functions. Because glial cells have been described as mediators of inflammatory processes in the brain, the existence of a causal link between hypothalamic inflammation and the deregulations of feeding behavior, leading to involuntary weight loss or obesity for example, has been suggested. Several inflammatory pathways that could impair the hypothalamic control of energy balance have been studied over the years such as, among others, toll-like receptors and canonical cytokines. Yet, less studied so far, chemokines also represent interesting candidates that could link the aforementioned pathways and the activity of hypothalamic neurons. Indeed, chemokines, in addition to their role in attracting immune cells to the inflamed site, have been suggested to be capable of neuromodulation. Thus, they could disrupt cellular activity together with synthesis and/or secretion of multiple neurotransmitters/mediators involved in the maintenance of energy balance. This review discusses the different inflammatory pathways that have been identified so far in the hypothalamus in the context of feeding behavior and body weight control impairments, with a particular focus on chemokines signaling that opens a new avenue in the understanding of the major role played by inflammation in obesity.

18.
C R Biol ; 329(8): 623-38; discussion 653-5, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16860280

RESUMO

A number of different neuropeptides exert powerful concerted controls on feeding behavior and energy balance, most of them being produced in hypothalamic neuronal networks under stimulation by anabolic and catabolic peripheral hormones such as ghrelin and leptin, respectively. These peptide-expressing neurons interconnect extensively to integrate the multiple opposing signals that mediate changes in energy expenditure. In the present review I have summarized our current knowledge about two key peptidic systems involved in regulating appetite and energy homeostasis, the melanocortin system (alpha-MSH, agouti and Agouti-related peptides, MC receptors and mahogany protein) and the melanin-concentrating hormone system (proMCH-derived peptides and MCH receptors) that contribute to satiety and feeding-initiation, respectively, with concurrent effects on energy expenditure. I have focused particularly on recent data concerning transgenic mice and the ongoing development of MC/MCH receptor antagonists/agonists that may represent promising drugs to treat human eating disorders on both sides of the energy balance (anorexia, obesity).


Assuntos
Metabolismo Energético/fisiologia , Comportamento Alimentar/fisiologia , Homeostase/fisiologia , Hormônios Hipotalâmicos/fisiologia , Melaninas/fisiologia , Hormônios Hipofisários/fisiologia , alfa-MSH/fisiologia , Proteína Agouti Sinalizadora , Animais , Hormônios/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Pró-Opiomelanocortina/fisiologia , Receptor Tipo 4 de Melanocortina/fisiologia , Transdução de Sinais
19.
Ann N Y Acad Sci ; 1351: 127-40, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26251227

RESUMO

Inflammation is an innate mechanism that defends organisms against harmful stimuli. Inflammation leads to the production and secretion of proinflammatory mediators that activate and recruit immune cells to damaged tissues, including the brain, to resolve the cause of inflammation. In the central nervous system, inflammation is referred to as neuroinflammation, which occurs in various pathological conditions of the brain. The primary role of neuroinflammation is to protect the brain. However, prolonged and/or inappropriate inflammation can be harmful for the brain, from individual cells to the whole tissue. This review focuses on a particular type of inflammatory mediator, chemokines, and describes their complex effects both under physiological and pathophysiological conditions of the brain. The clinical relevance of the multiple characters of chemokines is highlighted with respect to acute and chronic inflammation of the brain, including their actions in stroke and Alzheimer's disease, respectively.


Assuntos
Doença de Alzheimer/fisiopatologia , Encéfalo/imunologia , Quimiocinas/imunologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Acidente Vascular Cerebral/fisiopatologia , Doença de Alzheimer/imunologia , Encéfalo/citologia , Humanos , Hipóxia-Isquemia Encefálica/imunologia , Inflamação/imunologia , Mediadores da Inflamação/imunologia , Neuroimunomodulação/imunologia , Receptores de Quimiocinas/imunologia , Acidente Vascular Cerebral/imunologia
20.
Peptides ; 25(10): 1623-32, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15476929

RESUMO

A comparative analysis of the structure of the melanin-concentrating hormone (MCH) precursor reveals that this sequence has been subjected to a higher selection pressure in mammals than in teleosts, suggesting that the structural constraints have not been the same throughout the vertebrate lineage. In contrast, the MCH peptide sequence has been very well conserved in all species. A sensitive and reproducible eel skin assay was developed and allowed us to define the structural features needed for a full MCH bioactivity. It was shown that the minimal structure carrying the critical residues was the same in fishes and in mammals. A pharmacological approach confirmed that MCH receptor activation decreased the cAMP levels in the fish skin, but this effect appeared to be independent from a Galphai protein. We propose that one of the intracellular signaling pathways of the MCH receptor in fish skin is the activation of one or several cellular phosphodiesterases.


Assuntos
Hormônios Hipotalâmicos/química , Melaninas/química , Hormônios Hipofisários/química , Sequência de Aminoácidos , Animais , Sequência Conservada , Evolução Molecular , Peixes , Humanos , Hormônios Hipotalâmicos/genética , Mamíferos , Melaninas/genética , Dados de Sequência Molecular , Hormônios Hipofisários/genética , Reação em Cadeia da Polimerase/métodos , Primatas , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa