Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
1.
Biochem Biophys Res Commun ; 658: 18-26, 2023 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-37011479

RESUMO

Phenylbutyric acid (PBA) is a commonly used inhibitor of endoplasmic reticulum stress, as well as a histone deacetylase (HDAC) inhibitor, that increases hypothalamic expression of orexigenic neuropeptide Y (Npy). Elucidation of the dose-response relationship and mechanism of action of PBA may position this compound as a potential therapeutic for eating disorders where Npy is dysregulated, such as anorexia nervosa. The hypothalamic neuronal model mHypoE-41 was exposed to PBA (5 µM-5 mM) to assess the maximal Npy upregulation. Transcription factors and histone acetylation-related genes were assessed by qRT-PCR, as well as the involvement estrogen receptors (ER) using siRNA knockdown. Changes in global and Npy promoter-specific H3K9/14 acetylation were detected using western analysis and chromatin immunoprecipitation. Treatment with 5 mM PBA led to a 10-fold and 206-fold increase in Npy mRNA at 4 and 16 h, respectively, as well as increased NPY secretion. This induction was not observed with another orexigenic neuropeptide Agrp. PBA significantly increased the expression of Foxo1, Socs3 and Atf3 and the ERs Esr1 and Esr2 mRNA, but the PBA-mediated induction of Npy was not dependent on ERα or ERß. PBA induced histone H3K9/14 acetylation at 3 distinct Npy promoter regions, suggesting increased Npy transcriptional activation due to a more open chromatin structure. We also report changes in Hdac mRNAs by PBA and the fatty acid palmitate, highlighting the importance of epigenetic regulation in Npy transcription. Overall, we conclude that PBA has strong orexigenic potential and can robustly and specifically induce Npy in hypothalamic neurons through a mechanism likely involving histone H3 acetylation.


Assuntos
Histonas , Neuropeptídeo Y , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Histonas/metabolismo , Epigênese Genética , Acetilação , Hipotálamo/metabolismo , Neurônios/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
2.
J Neurochem ; 159(3): 574-589, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34482548

RESUMO

A contributing factor to the development of obesity is the consumption of a diet high in saturated fatty acids, such as palmitate. These fats induce hypothalamic neuroinflammation, which dysregulates neuronal function and induces orexigenic neuropeptide Y (Npy) to promote food intake. An inflammatory cytokine array identified multiple candidates that could mediate palmitate-induced up-regulation of Npy mRNA levels. Of these, visfatin or nicotinamide phosphoribosyltransferase (NAMPT), macrophage migratory inhibitory factor (MIF), and IL-17F were chosen for further study. Direct treatment of the neuropeptide Y/agouti-related peptide (NPY/AgRP)-expressing mHypoE-46 neuronal cell line with the aforementioned cytokines demonstrated that visfatin could directly induce Npy mRNA expression. Preventing the intracellular metabolism of palmitate through long-chain acyl-CoA synthetase (ACSL) inhibition was sufficient to block the palmitate-mediated increase in Npy gene expression. Furthermore, thin-layer chromatography revealed that in neurons, palmitate is readily incorporated into ceramides and defined species of phospholipids. Exogenous C16 ceramide, dipalmitoyl-phosphatidylcholine, and dipalmitoyl-phosphatidylethanolamine were sufficient to significantly induce Npy expression. This study suggests that the intracellular metabolism of palmitate and elevation of metabolites, including ceramide and phospholipids, are responsible for the palmitate-mediated induction of the potent orexigen Npy. Furthermore, this suggests that the regulation of Npy expression is less reliant on inflammatory cytokines per se than palmitate metabolites in a model of NPY/AgRP neurons. These lipid species likely induce detrimental downstream cellular signaling events ultimately causing an increase in feeding, resulting in an overweight phenotype and/or obesity.


Assuntos
Citocinas/farmacologia , Neuropeptídeo Y/biossíntese , Palmitatos/farmacologia , Acil Coenzima A/metabolismo , Animais , Linhagem Celular , Ceramidas/metabolismo , Meios de Cultivo Condicionados , Dieta Hiperlipídica , Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Nicotinamida Fosforribosiltransferase/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
3.
Neuroendocrinology ; 111(7): 678-695, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32575098

RESUMO

BACKGROUND: Bisphenol A (BPA) is a ubiquitous endocrine disrupting chemical and obesogen. Although limited evidence exists of the effects of BPA on hypothalamic agouti-related peptide (AgRP) levels, the mechanisms underlying these effects remain unknown. Given that AgRP is a potent orexigenic neuropeptide, determining the mechanism by which BPA increases AgRP is critical to preventing the progression to metabolic disease. METHODS: Using quantitative reverse transcriptase polymerase chain reaction, we investigated the response of Agrp-expressing mouse hypothalamic cell lines to BPA treatment. The percentage of total BPA entering hypothalamic cells in culture was quantified using an enzyme-linked immunosorbent assay. In order to identify the mechanism underlying BPA-mediated changes in Agrp, siRNA knockdown of transcription factors, FOXO1, CHOP, ATF3, ATF4, ATF6, and small-molecule inhibitors of endoplasmic reticulum stress, JNK or MEK/ERK were used. RESULTS: BPA increased mRNA levels of Agrp in six hypothalamic cell lines (mHypoA-59, mHypoE-41, mHypoA-2/12, mHypoE-46, mHypoE-44, and mHypoE-42). Interestingly, only 18% of the total BPA in the culture medium entered the cells after 24 h, suggesting that the exposure concentration is much lower than the treatment concentration. BPA increased pre-Agrp mRNA levels, indicating increased Agrp transcription. Knockdown of the transcription factor ATF3 prevented BPA-mediated increase in Agrp, pre-Agrp, and in part Npy mRNA levels. However, chemical chaperone, sodium phenylbutyrate, JNK inhibitor, SP600125, or the MEK/ERK inhibitor PD0352901 did not block BPA-induced Agrp upregulation. CONCLUSION: Overall, these results indicate that hypothalamic Agrp is susceptible to dysregulation by BPA and implicate ATF3 as a common mediator of the orexigenic effects of BPA in hypothalamic neurons.


Assuntos
Fator 3 Ativador da Transcrição/efeitos dos fármacos , Proteína Relacionada com Agouti/efeitos dos fármacos , Compostos Benzidrílicos/farmacologia , Disruptores Endócrinos/farmacologia , Estrogênios não Esteroides/farmacologia , Expressão Gênica/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fenóis/farmacologia , Animais , Células Cultivadas , Camundongos
4.
Diabetes Obes Metab ; 21(1): 61-72, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30062833

RESUMO

AIM: Omega-3 fatty acid ethyl ester supplements, available by prescription, are common in the treatment of dyslipidaemia in humans. Recent studies show that 3-carboxy-4-methyl-5-propyl-2-furanpropanoic acid (CMPF), a metabolite formed from fish oil supplementation, was able to prevent and reverse high fat diet (HFD)-induced fatty liver in mice. In the present study, we investigated the underlying molecular mechanisms responsible for CMPF's hepatic lipid-lowering effects. MATERIALS AND METHODS: CD1 male mice were i.p. injected with CMPF (dosage, 6 mg/kg) for 7 days, followed by 5 weeks of a 60% HFD to induce a fatty liver phenotype. Metabolic parameters, liver morphology, lipid content, protein expression and microarray analysis were assessed. We also utilized primary hepatocytes, an in vitro model, to further investigate the direct effects of CMPF on hepatic lipid utilization and biosynthesis. RESULTS: CMPF-treated mice display enhanced hepatic lipid clearance while hepatic lipid storage is prevented, thereby protecting against liver lipid accumulation and development of HFD-induced hepatic insulin resistance. Mechanistically, as CMPF enters the liver, it acts as an allosteric acetyl-coA carboxylase (ACC) inhibitor, which directly induces both fatty acid oxidation and hepatic production of fibroblast growth factor 21 (FGF21). A feed-back loop is initiated by CMPF, which exists between ACC inhibition, fatty acid oxidation and production of FGF21. As a consequence, an adaptive decrease in Insig2/SREBP-1c/FAS protein expression results in priming of the liver to prevent a HFD-induced fatty liver phenotype. CONCLUSION: CMPF is a potential driver of hepatic lipid metabolism, preventing diet-induced hepatic lipid deposition and insulin resistance in the long term.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Furanos/farmacologia , Resistência à Insulina/fisiologia , Fígado , Propionatos/farmacologia , Animais , Glicemia/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Fígado Gorduroso/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Metabolismo dos Lipídeos , Fígado/química , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos
5.
Am J Physiol Endocrinol Metab ; 315(2): E133-E140, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29631363

RESUMO

The brain, specifically the hypothalamus, controls whole body energy and glucose homeostasis through neurons that synthesize specific neuropeptides, whereas hypothalamic dysfunction is linked directly to insulin resistance, obesity, and type 2 diabetes mellitus. Nutrient excess, through overconsumption of a Western or high-fat diet, exposes the hypothalamus to high levels of free fatty acids, which induces neuroinflammation, endoplasmic reticulum stress, and dysregulation of neuropeptide synthesis. Furthermore, exposure to a high-fat diet also disrupts normal circadian rhythms, and conversely, clock gene knockout models have symptoms of metabolic disorders. While whole brain/animal studies have provided phenotypic end points and important clues to the genes involved, there are still major gaps in our understanding of the intracellular pathways and neuron-specific components that ultimately control circadian rhythms and energy homeostasis. Because of its complexity and heterogeneous nature, containing a diverse mix cell types, it is difficult to dissect the critical hypothalamic components involved in these processes. Of significance, we have the capacity to study these individual components using an extensive collection of both embryonic- and adult-derived, immortalized hypothalamic neuronal cell lines from rodents. These defined neuronal cell lines have been used to examine the impact of nutrient excess, such as palmitate, on circadian rhythms and neuroendocrine signaling pathways, as well as changes in vital neuropeptides, leading to the development of neuronal inflammation; the role of proinflammatory molecules in this process; and ultimately, restoration of normal signaling, clock gene expression, and neuropeptide synthesis in disrupted states by beneficial anti-inflammatory compounds in defined hypothalamic neurons.


Assuntos
Metabolismo Energético/fisiologia , Ácidos Graxos/fisiologia , Homeostase/fisiologia , Hipotálamo/fisiologia , Ácido Palmítico , Periodicidade , Animais , Humanos
6.
Neuroendocrinology ; 107(4): 387-399, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30352432

RESUMO

BACKGROUND AND OBJECTIVES: Elevated levels of saturated fatty acids (SFA) induce a state of neuroinflammation in the hypothalamus. It has been suggested that microglia sense palmitate, a prevalent circulating SFA, and act as mediators of this inflammatory process by communicating with neurons, particularly those involved in appetite regulation. In this study, we examined the inflammatory response to palmitate in immortalized microglial cell lines, BV-2 and IMG, and the subsequent effects on inflammatory gene expression in a model of NPY/AgRP neurons, mHypoE-46. METHODS: The BV-2 cells were treated with 50 µM palmitate for 4 and 24 h, and the transcriptional regulation of markers for inflammation and cellular stress was assessed using an RT2 Profiler PCR Array. Select genes were verified with qRT-PCR. The BV-2 and IMG cells were then co-cultured using 1.0-µm cell culture inserts with an immortalized hypothalamic cell line, mHypoE-46, to investigate potential intercellular communication between microglia and neurons. RESULTS: We found that palmitate increased the mRNA levels of specific inflammatory genes, and a general anti-inflammatory profile was revealed in the microglia cells. The mRNA changes in TNFα at 4 and 24 h in BV-2 cells were abrogated with the toll-like receptor 4 (TLR4) inhibitor, TAK-242, indicating the involvement of TLR4. Co-culture of mHypoE-46 neurons with microglia pre-treated with palmitate resulted in repression of TNFα expression in the hypothalamic neurons. As palmitate significantly increased IL-13 expression in microglia, the effect of this cytokine was tested in mHypoE-46 neurons. The addition of IL-13 to neuronal cultures normalized the palmitate-mediated increase in IL-6 and AgRP expression, suggesting that microglia may protect surrounding neurons, at least in part, through the release of IL-13. CONCLUSIONS: These results suggest a potential anti-inflammatory role of microglia towards the palmitate-induced neuroinflammation, and potentially energy homeostasis, in hypothalamic neurons.


Assuntos
Anti-Inflamatórios/farmacologia , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ácido Palmítico/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Técnicas de Cocultura , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/prevenção & controle , Lipopolissacarídeos/farmacologia , Camundongos , Microglia/fisiologia , Neurônios/fisiologia
7.
Acta Pharmacol Sin ; 39(5): 774-778, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29671415

RESUMO

Phoenixin (PNX) is a newly discovered peptide that has been linked to reproductive function, both in the hypothalamus and pituitary. This review will focus on the most recent discoveries related to this novel neuropeptide. Initially, it was found that PNX increased gonadotropin releasing hormone (GnRH)-stimulated luteinizing hormone (LH) release from pituitary cells. Importantly, knockdown of PNX in female rats extended the estrous cycle by 2.3 days. Using novel hypothalamic cell lines, we found that PNX has a stimulatory role on kisspeptin (Kiss) and GnRH gene expression and secretion. The PNX receptor was uncovered using siRNA knockdown of GPR173, an orphan receptor postulated to bind PNX. We have found that the PNX-R signaling through protein kinase A (PKA) in hypothalamic neurons. Althuogh a number of studies demonstrate that PNX plays an important role in reproductive function, there is also evidence that it may have other functions, regulating the heart, feeding, memory, and anxiety, both in the brain and the periphery.


Assuntos
Hormônios Hipotalâmicos/metabolismo , Hormônios Peptídicos/metabolismo , Peptídeos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Linhagem Celular , Comportamento Alimentar/fisiologia , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Hipotálamo/metabolismo , Masculino , Memória/fisiologia , Miocárdio/metabolismo , Medula Espinal/metabolismo
8.
Front Neuroendocrinol ; 36: 130-49, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25223866

RESUMO

Attesting to its intimate peripheral connections, hypothalamic neurons integrate nutritional and hormonal cues to effectively manage energy homeostasis according to the overall status of the system. Extensive progress in the identification of essential transcriptional and post-translational mechanisms regulating the controlled expression and actions of hypothalamic neuropeptides has been identified through the use of animal and cell models. This review will introduce the basic techniques of hypothalamic investigation both in vivo and in vitro and will briefly highlight the key advantages and challenges of their use. Further emphasis will be place on the use of immortalized models of hypothalamic neurons for in vitro study of feeding regulation, with a particular focus on cell lines proving themselves most fruitful in deciphering fundamental basics of NPY/AgRP, Proglucagon, and POMC neuropeptide function.


Assuntos
Metabolismo Energético/fisiologia , Homeostase/fisiologia , Hipotálamo/fisiologia , Neurônios/fisiologia , Animais , Linhagem Celular , Hipotálamo/citologia , Neurônios/citologia , Neuropeptídeos/fisiologia
9.
Am J Physiol Regul Integr Comp Physiol ; 311(2): R217-21, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27306829

RESUMO

The hypothalamus is responsible for the control of many of our physiological responses, including energy homeostasis. Of interest, there are a number of instances of sexual dimorphism documented with regard to metabolic processes. This review will discuss the necessity of utilizing both male and female models when studying the mechanisms underlying energy homeostasis, particularly those originating at the level of the hypothalamus. Because obesity often results in central neuroinflammation, we describe markers that could be used to study differences between male and female models, both the whole organism and also at the cellular level. Our laboratory has generated a wide array of immortalized hypothalamic cell models, originating from male and female rodents that we suggest could be beneficial for these types of studies. It is imperative that both sexes are considered before any recommendations for therapeutic interventions are considered.


Assuntos
Regulação do Apetite/imunologia , Hipotálamo/imunologia , Inflamação Neurogênica/imunologia , Neurônios/imunologia , Neuropeptídeos/imunologia , Animais , Linhagem Celular , Feminino , Humanos , Masculino , Camundongos , Modelos Imunológicos , Caracteres Sexuais
10.
Am J Physiol Regul Integr Comp Physiol ; 308(5): R337-50, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25519730

RESUMO

The suprachiasmatic nucleus (SCN) in the mammalian hypothalamus functions as an endogenous pacemaker that generates and maintains circadian rhythms throughout the body. Next to this central clock, peripheral oscillators exist in almost all mammalian tissues. Whereas the SCN is mainly entrained to the environment by light, peripheral clocks are entrained by various factors, of which feeding/fasting is the most important. Desynchronization between the central and peripheral clocks by, for instance, altered timing of food intake can lead to uncoupling of peripheral clocks from the central pacemaker and is, in humans, related to the development of metabolic disorders, including obesity and Type 2 diabetes. Diets high in fat or sugar have been shown to alter circadian clock function. This review discusses the recent findings concerning the influence of nutrients, in particular fatty acids and glucose, on behavioral and molecular circadian rhythms and will summarize critical studies describing putative mechanisms by which these nutrients are able to alter normal circadian rhythmicity, in the SCN, in non-SCN brain areas, as well as in peripheral organs. As the effects of fat and sugar on the clock could be through alterations in energy status, the role of specific nutrient sensors will be outlined, as well as the molecular studies linking these components to metabolism. Understanding the impact of specific macronutrients on the circadian clock will allow for guidance toward the composition and timing of meals optimal for physiological health, as well as putative therapeutic targets to regulate the molecular clock.


Assuntos
Relógios Biológicos , Ritmo Circadiano , Dieta , Ingestão de Alimentos , Jejum/metabolismo , Núcleo Supraquiasmático/metabolismo , Animais , Transtornos Cronobiológicos/metabolismo , Transtornos Cronobiológicos/fisiopatologia , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/metabolismo , Dieta/efeitos adversos , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/metabolismo , Sacarose Alimentar/efeitos adversos , Sacarose Alimentar/metabolismo , Metabolismo Energético , Humanos , Estado Nutricional , Transdução de Sinais , Núcleo Supraquiasmático/fisiopatologia
11.
Am J Physiol Endocrinol Metab ; 306(12): E1341-53, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24760992

RESUMO

Chronic low-grade inflammation is an important contributor to the development of insulin resistance, a hallmark of type 2 diabetes mellitus (T2DM). Obesity and high-fat feeding lead to infiltration of immune cells into metabolic tissues, promoting inflammation and insulin resistance. We hypothesized that macrophages from mice lacking NOX2 (Cybb), an essential component of the NADPH oxidase complex highly expressed in immune cells and associated with their inflammatory response, would be less inflammatory and that these mice would be protected from the development of high-fat-induced insulin resistance. Bone marrow-derived macrophages from NOX2 knockout (NOX2-KO) mice expressed lower levels of inflammatory markers (Nos2, Il6); however, NOX2-KO mice were hyperphagic and gained more weight than wild-type (WT) mice when fed either a chow or a high-fat (HF) diet. Surprisingly, NOX2-KO mice stored less lipid in epididymal white adipose tissue but more lipid in liver and had higher indexes of liver inflammation and macrophage infiltration than WT mice. Contrary to our hypothesis, HF-fed NOX2-KO mice were hyperinsulinemic and more insulin resistant than HF-fed WT mice, likely as a result of their higher hepatic steatosis and inflammation. In summary, NOX2 depletion promoted hyperphagia, hepatic steatosis, and inflammation with either normal or high-fat feeding, exacerbating insulin resistance. We propose that NOX2 participates in food intake control and lipid distribution in mice.


Assuntos
Regulação do Apetite , Fígado Gorduroso/etiologia , Hiperfagia/metabolismo , Resistência à Insulina , Metabolismo dos Lipídeos , Fígado/metabolismo , Glicoproteínas de Membrana/metabolismo , NADPH Oxidases/metabolismo , Tecido Adiposo Branco/imunologia , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/patologia , Animais , Comportamento Apetitivo , Comportamento Animal , Células Cultivadas , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/etiologia , Dieta Hiperlipídica/efeitos adversos , Fígado Gorduroso/complicações , Hiperfagia/imunologia , Hiperfagia/patologia , Hiperfagia/fisiopatologia , Fígado/imunologia , Fígado/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidase 2 , NADPH Oxidases/genética , Hepatopatia Gordurosa não Alcoólica , Obesidade/complicações , Obesidade/etiologia
12.
J Neuroinflammation ; 11: 60, 2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24674717

RESUMO

BACKGROUND: Overnutrition and the ensuing hypothalamic inflammation is a major perpetuating factor in the development of metabolic diseases, such as obesity and diabetes. Inflamed neurons of the CNS fail to properly regulate energy homeostasis leading to pathogenic changes in glucose handling, feeding, and body weight. Hypothalamic neurons are particularly sensitive to pro-inflammatory signals derived locally and peripherally, and it is these neurons that become inflamed first upon high fat feeding. Given the prevalence of metabolic disease, efforts are underway to identify therapeutic targets for this inflammatory state. At least in the periphery, omega-3 fatty acids and their receptor, G-protein coupled receptor 120 (GPR120), have emerged as putative targets. The role for GPR120 in the hypothalamus or CNS in general is poorly understood. METHODS: Here we introduce a novel, immortalized cell model derived from the rat hypothalamus, rHypoE-7, to study GPR120 activation at the level of the individual neuron. Gene expression levels of pro-inflammatory cytokines were studied by quantitative reverse transcriptase-PCR (qRT-PCR) upon exposure to tumor necrosis factor α (TNFα) treatment in the presence or absence of the polyunsaturated omega-3 fatty acid docosahexaenoic acid (DHA). Signal transduction pathway involvement was also studied using phospho-specific antibodies to key proteins by western blot analysis. RESULTS: Importantly, rHypoE-7 cells exhibit a transcriptional and translational inflammatory response upon exposure to TNFα and express abundant levels of GPR120, which is functionally responsive to DHA. DHA pretreatment prevents the inflammatory state and this effect was inhibited by the reduction of endogenous GPR120 levels. GPR120 activates both AKT (protein kinase b) and ERK (extracellular signal-regulated kinase); however, the anti-inflammatory action of this omega-3 fatty acid (FA) receptor is AKT- and ERK-independent and likely involves the GPR120-transforming growth factor-ß-activated kinase 1 binding protein (TAB1) interaction as identified in the periphery. CONCLUSIONS: Taken together, GPR120 is functionally active in the hypothalamic neuronal line, rHypoE-7, wherein it mediates the anti-inflammatory actions of DHA to reduce the inflammatory response to TNFα.


Assuntos
Anti-Inflamatórios/farmacologia , Neurônios/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Animais , Linhagem Celular Transformada , Ácidos Docosa-Hexaenoicos/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Hipotálamo/citologia , Proteínas I-kappa B/metabolismo , Imunoprecipitação , Metilaminas/farmacologia , Propionatos/farmacologia , Piridinas/farmacologia , RNA Interferente Pequeno/farmacologia , Ratos , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia
13.
Am J Physiol Regul Integr Comp Physiol ; 307(8): R1049-60, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25144192

RESUMO

Diets high in saturated fatty acids (SFAs) are associated with the development of circadian dysregulation, obesity, and Type 2 diabetes mellitus. Conversely, polyunsaturated fatty acids (PUFAs) have recently been identified to improve insulin sensitivity, reduce weight gain, and relieve obesity-induced inflammation. While saturated fatty acids, such as the prevalent dietary fatty acid palmitate, have been implicated in circadian disruption, there is a paucity of studies regarding the effects of PUFAs on circadian parameters. Therefore, the immortalized murine neuronal model, mHypoE-37, was utilized to examine the effects of the SFA palmitate and omega-3 PUFA docosahexaenoic acid (DHA) on circadian rhythms. The mHypoE-37 neurons express the core clock genes, Bmal1, Per2, and Rev-erbα, in a circadian manner. 25 µM of palmitate significantly increased the transcriptional expression of Bmal1, without altering the expression of inflammatory markers TLR4, IκBα, and IL-6, nor the orexigenic neuropeptide AgRP, suggesting that the observed disruption of the molecular clock is the result of a mechanism distinct from that of hypothalamic cellular inflammation. Furthermore, treatment with the PUFA DHA resulted in alterations in the circadian expression profile of Bmal1, although differentially from the effects of palmitate. In the presence of DHA, the disruptive effects of palmitate on Bmal1 were less pronounced, suggesting a protective effect of DHA. These studies are the first to identify the potential for omega-3 PUFAs to protect against palmitate-mediated dysregulation of circadian parameters and will ultimately improve the understanding of circadian control mechanisms.


Assuntos
Relógios Circadianos/genética , Ritmo Circadiano/efeitos dos fármacos , Ácidos Docosa-Hexaenoicos/farmacologia , Hipotálamo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Palmitatos/farmacologia , Transcriptoma/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Ritmo Circadiano/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Hipotálamo/citologia , Hipotálamo/metabolismo , Camundongos , Modelos Animais , Neurônios/citologia , Neurônios/metabolismo , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Fosforilação , Transcriptoma/genética
14.
Biomedicines ; 12(4)2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38672211

RESUMO

Cellular microRNAs (miRNAs) can be selectively secreted or retained, adding another layer to their critical role in regulating human health and disease. To date, select RNA-binding proteins (RBPs) have been proposed to be a mechanism underlying miRNA localization, but the overall relevance of RBPs in systematic miRNA sorting remains unclear. This study profiles intracellular and small extracellular vesicles' (sEVs) miRNAs in NPY-expressing hypothalamic neurons. These findings were corroborated by the publicly available sEV and intracellular miRNA profiles of white and brown adipocytes, endothelium, liver, and muscle from various databases. Using experimentally determined binding motifs of 93 RBPs, our enrichment analysis revealed that sEV-originating miRNAs contained significantly different RBP motifs than those of intracellularly retained miRNAs. Multiple RBP motifs were shared across cell types; for instance, RBM4 and SAMD4 are significantly enriched in neurons, hepatocytes, skeletal muscle, and endothelial cells. Homologs of both proteins physically interact with Argonaute1/2 proteins, suggesting that they play a role in miRNA sorting. Machine learning modelling also demonstrates that significantly enriched RBP motifs could predict cell-specific preferential miRNA sorting. Non-optimized machine learning modeling of the motifs using Random Forest and Naive Bayes in all cell types except WAT achieved an area under the receiver operating characteristic (ROC) curve of 0.77-0.84, indicating a high predictive accuracy. Given that the RBP motifs have a significant predictive power, these results underscore the critical role that RBPs play in miRNA sorting within mammalian cells and reinforce the importance of miRNA sequencing in preferential localization. For the future development of small RNA therapeutics, considering these RBP-RNA interactions could be crucial to maximize delivery effectiveness and minimize off-target effects.

15.
Genes (Basel) ; 15(4)2024 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-38674332

RESUMO

Diets high in saturated fatty acids are associated with obesity and infertility. Palmitate, the most prevalent circulating saturated fatty acid, is sensed by hypothalamic neurons, contributing to homeostatic dysregulation. Notably, palmitate elevates the mRNA levels of gonadotropin-releasing hormone (Gnrh) mRNA and its activating transcription factor, GATA binding protein 4 (Gata4). GATA4 is essential for basal Gnrh expression by binding to its enhancer region, with Oct-1 (Oct1) and CEBP-ß (Cebpb) playing regulatory roles. The pre- and post-transcriptional control of Gnrh by palmitate have not been investigated. Given the ability of palmitate to alter microRNAs (miRNAs), we hypothesized that palmitate-mediated dysregulation of Gnrh mRNA involves specific miRNAs. In the mHypoA-GnRH/GFP neurons, palmitate significantly downregulated six miRNAs (miR-125a, miR-181b, miR-340, miR-351, miR-466c and miR-503), and the repression was attenuated by co-treatment with 100 µM of oleate. Subsequent mimic transfections revealed that miR-466c significantly downregulates Gnrh, Gata4, and Chop mRNA and increases Per2, whereas miR-340 upregulates Gnrh, Gata4, Oct1, Cebpb, and Per2 mRNA. Our findings suggest that palmitate may indirectly regulate Gnrh at both the pre- and post-transcriptional levels by altering miR-466c and miR-340, which in turn regulate transcription factor expression levels. In summary, palmitate-mediated dysregulation of Gnrh and, consequently, reproductive function involves parallel transcriptional mechanisms.


Assuntos
Regulação da Expressão Gênica , Hormônio Liberador de Gonadotropina , MicroRNAs , Palmitatos , MicroRNAs/genética , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Animais , Palmitatos/metabolismo , Camundongos , Regulação da Expressão Gênica/efeitos dos fármacos , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Hipotálamo/metabolismo
16.
Mol Cell Endocrinol ; 586: 112179, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38387703

RESUMO

Neuropeptide Y (Npy) is an abundant neuropeptide expressed in the central and peripheral nervous systems. NPY-secreting neurons in the hypothalamic arcuate nucleus regulate energy homeostasis, and Npy mRNA expression is regulated by peripheral nutrient and hormonal signals like leptin, interleukin-6 (IL-6), and fatty acids. This study demonstrates that IL-6, which phosphorylates tyrosine 705 (Y705) of STAT3, decreased Npy mRNA in arcuate immortalized hypothalamic neurons. In parallel, inhibitors of STAT3-Y705 phosphorylation, stattic and cucurbitacin I, robustly upregulated Npy mRNA. Chromatin-immunoprecipitation showed high baseline total STAT3 binding to multiple regulatory regions of the Npy gene, which are decreased by IL-6 exposure. The STAT3-Npy interaction was further examined in obesity-related pathologies. Notably, in four different hypothalamic neuronal models where palmitate potently stimulated Npy mRNA, Socs3, a specific STAT3 activity marker, was downregulated and was negatively correlated with Npy mRNA levels (R2 = 0.40, p < 0.001), suggesting that disrupted STAT3 signaling is involved in lipotoxicity-mediated dysregulation of Npy. Finally, human NPY SNPs that map to human obesity or body mass index were investigated for potential STAT3 binding sites. Although none of the SNPs were linked to direct STAT3 binding, analysis show that rs17149106 (-602 G > T) is located on an upstream enhancer element of NPY, where the variant is predicted to disrupt validated binding of KLF4, a known inhibitory cofactor of STAT3 and downstream effector of leptin signaling. Collectively, this study demonstrates that STAT3 signaling negatively regulates Npy transcription, and that disruption of this interaction may contribute to metabolic disorders.


Assuntos
Leptina , Neuropeptídeo Y , Humanos , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Leptina/farmacologia , Leptina/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Hipotálamo/metabolismo , Obesidade/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Neurônios/metabolismo , RNA Mensageiro/genética , Fator de Transcrição STAT3/metabolismo
17.
Neuroscience ; 551: 43-54, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38788830

RESUMO

The generation of astrocyte cell lines from the hypothalamus is key to study glial involvement in hypothalamic physiology, including energy homeostasis. As such, we immortalized astrocytes from the hypothalamus of an adult male CD-1 mouse using SV40 T-antigen to generate the mHypoA-Ast1 cell line. A comparative approach was taken with two other murine GFAP-expressing cell lines that were also generated in this study: a mixed glial cell line from the cortex (mCortA-G1) and an oligodendrocyte cell line from the brainstem (mBstA-Olig1), as well as an established microglial cell line (IMG). mHypoA-Ast1 cells express GFAP, alongside other astrocytic markers such as Aldh1l1, Aqp4, Glt1 and S100b, and express low levels of microglial, ependymal and oligodendrocyte markers. 100 ng/mL lipopolysaccharide (LPS) elevated mRNA levels of Il6, Il1b, Tnfα and Cxcl5 in mHypoA-Ast1 cells after 4 h, while 50 µM palmitate increased Il6 and Chop mRNA, demonstrating the ability of these cells to respond to inflammatory and nutrient signals. Interestingly, co-culture of mHypoA-Ast1 cells with mHypoE-N46 hypothalamic neuronal cells prevented the palmitate-mediated increase in orexigenic neuropeptide Agrp mRNA in mHypoE-N46 cells, suggesting that this cell line can alter neuronal responses to nutrients. In conclusion, we report mHypoA-Ast1 cells representing a functional astrocyte cell line from the adult mouse brain that can be used to study the complex interactions of hypothalamic cells, as well as dysregulation that may occur in disease states, providing a key tool for neuroendocrine research.

18.
J Neuroendocrinol ; : e13398, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38733120

RESUMO

Phoenixin (PNX) is a conserved secreted peptide that was identified 10 years ago with numerous studies published on its pleiotropic functions. PNX is associated with estrous cycle length, protection from a high-fat diet, and reduction of anxiety behavior. However, no study had yet evaluated the impact of deleting PNX in the whole animal. We sought to evaluate a mouse model lacking the PNX parent gene, small integral membrane protein 20 (Smim20), and the resulting effect on reproduction, energy homeostasis, and anxiety. We found that the Smim20 knockout mice had normal fertility and estrous cycle lengths. Consistent with normal fertility, the hypothalamii of the knockout mice showed no changes in the levels of reproduction-related genes, but the male mice had some changes in energy homeostasis-related genes, such as melanocortin receptor 4 (Mc4r). When placed on a high-fat diet, the wildtype and knockout mice responded similarly, but the male heterozygous mice gained slightly less weight. When placed in an open field test box, the female knockout mice traveled less distance in the outer zone, indicating alterations in anxiety or locomotor behavior. In summary, the homozygous knockout of PNX did not alter fertility and modestly alters a few neuroendocrine genes in response to a high-fat diet, especially in the female mice. However, it altered the behavior of mice in an open field test. PNX therefore may not be crucial for reproductive function or weight, however, we cannot rule out possible compensatory mechanisms in the knockout model. Understanding the role of PNX in physiology may ultimately lead to an enhanced understanding of neuroendocrine mechanisms involving this enigmatic peptide.

19.
Aging Cell ; 23(2): e14047, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37994388

RESUMO

Orexigenic neurons expressing agouti-related protein (AgRP) and neuropeptide Y in the arcuate nucleus (ARC) of the hypothalamus are activated in response to dynamic variations in the metabolic state, including exercise. We previously observed that carnitine palmitoyltransferase 1a (CPT1A), a rate-limiting enzyme of mitochondrial fatty acid oxidation, is a key factor in AgRP neurons, modulating whole-body energy balance and fluid homeostasis. However, the effect of CPT1A in AgRP neurons in aged mice and during exercise has not been explored yet. We have evaluated the physical and cognitive capacity of adult and aged mutant male mice lacking Cpt1a in AgRP neurons (Cpt1a KO). Adult Cpt1a KO male mice exhibited enhanced endurance performance, motor coordination, locomotion, and exploration compared with control mice. No changes were observed in anxiety-related behavior, cognition, and muscle strength. Adult Cpt1a KO mice showed a reduction in gastrocnemius and tibialis anterior muscle mass. The cross-sectional area (CSA) of these muscles were smaller than those of control mice displaying a myofiber remodeling from type II to type I fibers. In aged mice, changes in myofiber remodeling were maintained in Cpt1a KO mice, avoiding loss of physical capacity during aging progression. Additionally, aged Cpt1a KO mice revealed better cognitive skills, reduced inflammation, and oxidative stress in the hypothalamus and hippocampus. In conclusion, CPT1A in AgRP neurons appears to modulate health and protects against aging. Future studies are required to clarify whether CPT1A is a potential antiaging candidate for treating diseases affecting memory and physical activity.


Assuntos
Carnitina O-Palmitoiltransferase , Envelhecimento Saudável , Animais , Masculino , Camundongos , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Carnitina O-Palmitoiltransferase/genética , Carnitina O-Palmitoiltransferase/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo
20.
Brain Res ; 1810: 148367, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37054963

RESUMO

Exosomes (sEVs) are extracellular vesicles involved in the pathogenesis of obesity. Notably, exosomal microRNAs (miRNAs) have emerged as crucial mediators of communication between cells and are involved in the development of obesity. One region of the brain known to be dysregulated in obesity is the hypothalamus. It coordinates whole-body energy homeostasis through stimulation and inhibition of the orexigenic neuropeptide (NPY)/agouti-related peptide (AgRP) neurons and anorexigenic proopiomelanocortin (POMC) neurons. A role for hypothalamic astrocytic exosomes in communication with POMC neurons was previously elucidated. Yet, it was unknown whether NPY/AgRP neurons secreted exosomes. We previously established that the saturated fat palmitate alters the intracellular levels of miRNAs and we now questioned whether palmitate would also alter the miRNA content of exosomal miRNAs. We found that the mHypoE-46 cell line secreted particles consistent with the size of exosomes and that palmitate altered levels of a spectrum of miRNAs associated with exosomes. The predicted KEGG pathways of the collective miRNA predicted targets included fatty acid metabolism and type II diabetes mellitus. Of note, one of these altered secreted miRNAs was miR-2137, which was also altered within the cells. We also found that while sEVs collected from the mHypoE-46 neurons increased Pomc mRNA in the mHypoA-POMC/GFP-2 cells after 48 h, the effect was absent with sEVs isolated following palmitate treatment, indicating another potential route by which palmitate promotes obesity. Hypothalamic neuronal exosomes may therefore play a role in the control of energy homeostasis that may be disrupted in obese conditions.


Assuntos
Diabetes Mellitus Tipo 2 , Vesículas Extracelulares , Palmitatos , Humanos , Proteína Relacionada com Agouti/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Vesículas Extracelulares/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo , Obesidade/metabolismo , Palmitatos/farmacologia , Palmitatos/metabolismo , Pró-Opiomelanocortina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA