Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Ann Neurol ; 95(4): 665-676, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38379184

RESUMO

OBJECTIVE: To examine associations of serum insulin and related measures with neuropathology and cognition in older persons. METHODS: We studied 192 older persons (96 with diabetes and 96 without, matched by sex and balanced by age-at-death, education, and postmortem interval) from a community-based, clinical-pathologic study of aging, with annual evaluations including neuropsychological testing (summarized into global cognition and 5 cognitive domains) and postmortem autopsy. We assessed serum insulin, glucose, leptin, adiponectin, hemoglobin A1C, advanced glycation-end products (AGEs), and receptors for advanced glycation-end products, and calculated the Homeostasis Model Assessment of Insulin Resistance (HOMA-IR) and adiponectin-to-leptin ratio. Using adjusted regression analyses, we examined the associations of serum measures with neuropathology of cerebrovascular disease and Alzheimer's disease, and with the level of cognition proximate-to-death. RESULTS: Higher HOMA-IR was associated with the presence of brain infarcts and specifically microinfarcts, and higher HOMA-IR and leptin were each associated with subcortical infarcts. Further, higher leptin levels and lower adiponectin-to-leptin ratios were associated with the presence of moderate-to-severe atherosclerosis. Serum insulin and related measures were not associated with the level of Alzheimer's disease pathology, as assessed by global, as well as amyloid burden or tau tangle density scores. Regarding cognitive outcomes, higher insulin and leptin levels, and lower adiponectin and receptors for advanced glycation-end products levels, respectively, were each associated with lower levels of global cognition. INTERPRETATION: Peripheral insulin resistance indicated by HOMA-IR and related serum measures was associated with a greater burden of cerebrovascular neuropathology and lower cognition. ANN NEUROL 2024;95:665-676.


Assuntos
Doença de Alzheimer , Diabetes Mellitus , Resistência à Insulina , Doenças do Sistema Nervoso , Humanos , Idoso , Idoso de 80 Anos ou mais , Leptina , Doença de Alzheimer/patologia , Adiponectina , Cognição , Insulina
2.
Cell Rep ; 42(12): 113512, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-38039129

RESUMO

Mismatch between CO2 production (Vco2) and respiration underlies the pathogenesis of obesity hypoventilation. Leptin-mediated CNS pathways stimulate both metabolism and breathing, but interactions between these functions remain elusive. We hypothesized that LEPRb+ neurons of the dorsomedial hypothalamus (DMH) regulate metabolism and breathing in obesity. In diet-induced obese LeprbCre mice, chemogenetic activation of LEPRb+ DMH neurons increases minute ventilation (Ve) during sleep, the hypercapnic ventilatory response, Vco2, and Ve/Vco2, indicating that breathing is stimulated out of proportion to metabolism. The effects of chemogenetic activation are abolished by a serotonin blocker. Optogenetic stimulation of the LEPRb+ DMH neurons evokes excitatory postsynaptic currents in downstream serotonergic neurons of the dorsal raphe (DR). Administration of retrograde AAV harboring Cre-dependent caspase to the DR deletes LEPRb+ DMH neurons and abolishes metabolic and respiratory responses to leptin. These findings indicate that LEPRb+ DMH neurons match breathing to metabolism through serotonergic pathways to prevent obesity-induced hypoventilation.


Assuntos
Hipoventilação , Leptina , Camundongos , Animais , Leptina/metabolismo , Hipoventilação/metabolismo , Obesidade/metabolismo , Respiração , Hipotálamo/metabolismo , Receptores para Leptina/metabolismo
3.
Clin Med Insights Endocrinol Diabetes ; 16: 11795514231218592, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38107873

RESUMO

Background: The rapidly rising cardiometabolic disease (CMD) burden in urbanizing sub-Saharan African populations and among sub-Saharan African migrants in Europe likely affects serum adiponectin and leptin levels, but this has not yet been quantified. Objectives: To compare the serum levels of adiponectin and leptin among migrant, and non-migrant (urban and rural) populations of Ghanaian descent. Methods: Cross-sectional analysis of serum leptin and adiponectin in the multi-centre Research on Obesity and Diabetes among African Migrants (RODAM) study. Logistic-regression models were used to examine the association between these adipocyte-derived hormones after stratification (sex, geographic area) and adjustments for potential confounders. Results: A total of 2518 Ghanaians were included. Rural participants had the highest serum adiponectin and lowest leptin levels compared to Amsterdam and urban Ghanaians (P < .001). In fully adjusted models, participants living in urban Ghana had significantly higher odds of hyperleptinemia compared to rural participants (women-odds ratio 2.88; 95% CI, 1.12-7.38, P = .028 and men 43.52, 95% CI, 4.84-391.25, P < .001). Urban Ghanaian men also had higher odds of elevated leptin: adiponectin ratio (6.29, 95% CI, 1.43-27.62, P = .015). The odds of hyperleptinemia were only higher in Amsterdam Ghanaian men (10.56; 95% CI, 1.11-100.85, P = .041), but not in women (0.85; 95% CI, 0.30-2.41, P = .759). There was no significant association between hypoadiponectinemia and geographical location in both sexes. Conclusion: Urbanization is associated with serum adiponectin and leptin levels after adjusting for confounding covariates in sub-Saharan Africans. These findings serve as a backdrop for further research on the role adipokines play in CMD epidemiology among Africans.

4.
Sleep ; 46(8)2023 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-37262435

RESUMO

Obesity and male sex are main risk factors for sleep-disordered breathing (SDB). We have shown that male diet-induced obesity (DIO) mice develop hypoventilation, sleep apnea, and sleep fragmentation. The effects of DIO on breathing and sleep architecture in females have not been investigated. We hypothesized that female mice are less susceptible to the detrimental effects of DIO on sleep and SDB compared to males. Female DIO-C57BL/6J and lean C57BL/6J mice underwent 24-hour metabolic studies and were exposed to 8% CO2 to measure the hypercapnic ventilatory response (HCVR), and sleep studies. Ventilatory response to arousals was calculated as ratio of the average and peak minute ventilation (VE) during each arousal relative to the baseline VE. Breathing stability was measured with Poincaré plots of VE. Female obesity was associated with decreased metabolism, indicated by reduced oxygen consumption (VO2) and CO2 production (VCO2). VE in 8% CO2 and HCVR were significantly attenuated during wakefulness. NREM sleep duration was reduced in DIO mice, but REM sleep was preserved. Ventilation during NREM and REM sleep was augmented compared to lean mice. Arousal frequency was similar between groups. Obesity increased the frequency of spontaneous arousals, whereas the apnea index was 4-fold reduced in DIO compared to lean mice. Obesity decreased pre- and post-apnea arousals. Obese mice had more stable breathing with reduced ventilatory response to arousals, compared to lean females. We conclude that obese female mice are protected against SDB, which appears to be related to an attenuated CO2 responsiveness, compared to the lean state.


Assuntos
Dióxido de Carbono , Síndromes da Apneia do Sono , Feminino , Masculino , Animais , Camundongos , Camundongos Endogâmicos C57BL , Dieta , Obesidade/complicações , Sono , Síndromes da Apneia do Sono/complicações , Hipercapnia
6.
J Physiol ; 600(23): 5145-5162, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36214387

RESUMO

Sleep-disordered breathing (SDB) affects over 50% of obese individuals. Exaggerated hypoxic chemoreflex is a cardinal trait of SDB in obesity. We have shown that leptin acts in the carotid bodies (CB) to augment chemoreflex and that leptin activates the transient receptor potential melastatin 7 (TRPM7) channel. However, the effect of leptin-TRPM7 signalling in CB on breathing and SDB has not been characterized in diet-induced obesity (DIO). We hypothesized that leptin acts via TRPM7 in the CB to increase chemoreflex leading to SDB in obesity. DIO mice were implanted with EEG/EMG electrodes and transfected with Leprb short hairpin RNA (shRNA) or Trpm7 shRNA vs. control shRNA in the CB area bilaterally. Mice underwent a full-polysomnography and metabolic studies at baseline and after transfection. Ventilatory responses to hypoxia and hypercapnia were assessed during wakefulness. Leprb and Trpm7 were upregulated and their promoters were demethylated in the CB of DIO mice. Leprb knockdown in the CB did not significantly affect ventilation. Trpm7 knockdown in the CB stimulated breathing during sleep in normoxia. These effects were not driven by changes in CB chemosensitivity or metabolism. Under sustained hypoxia, Trpm7 shRNA in the CB augmented ventilation during sleep, but decreased oxyhaemoglobin saturation. We conclude that the suppression of TRPM7 in the CB improved sleep-related hypoventilation and that the respiratory effects of CB TRPM7 channels in obesity are independent of leptin. TRPM7 signalling in the CB could be a therapeutic target for the treatment of obesity-related SDB. KEY POINTS: The leptin-TRPM7 axis in the carotid bodies may play an important role in the pathogenesis of sleep-disordered breathing. TRPM7 channels regulate breathing during sleep by acting peripherally in the carotid bodies. Suppression of TRPM7 signalling in the carotid bodies improves the obesity-induced hypoventilation in mice. Pharmacological blockade of TRPM7 channels in the carotid bodies could be a therapy for sleep-disordered breathing in obesity.


Assuntos
Corpo Carotídeo , Síndromes da Apneia do Sono , Canais de Cátion TRPM , Canais de Potencial de Receptor Transitório , Camundongos , Animais , Corpo Carotídeo/fisiologia , Leptina/metabolismo , Hipoventilação/metabolismo , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , RNA Interferente Pequeno , Sono/fisiologia , Obesidade/complicações , Obesidade/metabolismo , Camundongos Obesos , Síndromes da Apneia do Sono/metabolismo , Hipóxia/complicações , Hipóxia/metabolismo
7.
J Cell Physiol ; 237(8): 3421-3432, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35822903

RESUMO

Hepatic glucose production (HGP) is crucial for the maintenance of normal glucose homeostasis. Although hepatic insulin resistance contributes to excessive glucose production, its mechanism is not well understood. Here, we show that inositol polyphosphate multikinase (IPMK), a key enzyme in inositol polyphosphate biosynthesis, plays a role in regulating hepatic insulin signaling and gluconeogenesis both in vitro and in vivo. IPMK-deficient hepatocytes exhibit decreased insulin-induced activation of Akt-FoxO1 signaling. The expression of messenger RNA levels of phosphoenolpyruvate carboxykinase 1 (Pck1) and glucose 6-phosphatase (G6pc), key enzymes mediating gluconeogenesis, are increased in IPMK-deficient hepatocytes compared to wild type hepatocytes. Importantly, re-expressing IPMK restores insulin sensitivity and alleviates glucose production in IPMK-deficient hepatocytes. Moreover, hepatocyte-specific IPMK deletion exacerbates hyperglycemia and insulin sensitivity in mice fed a high-fat diet, accompanied by an increase in HGP during pyruvate tolerance test and reduction in Akt phosphorylation in IPMK deficient liver. Our results demonstrate that IPMK mediates insulin signaling and gluconeogenesis and may be potentially targeted for treatment of diabetes.


Assuntos
Glucose , Resistência à Insulina , Insulina , Fígado , Fosfotransferases (Aceptor do Grupo Álcool) , Animais , Proteína Forkhead Box O1/metabolismo , Glucose/metabolismo , Glucose-6-Fosfatase/metabolismo , Hepatócitos/metabolismo , Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fígado/metabolismo , Camundongos , Fosfoenolpiruvato Carboxiquinase (GTP)/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
8.
J Infect Dis ; 226(9): 1626-1636, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-35512127

RESUMO

BACKGROUND: Antiretroviral therapy (ART) containing integrase strand transfer inhibitors (INSTIs) has been associated with weight gain in both ART initiation and switch studies, especially in women, but the underlying mechanisms are unclear. METHODS: The effects of dolutegravir (DTG) on food intake, energy expenditure, oxygen consumption in female mice, and gene expression from adipose tissues were assessed. Human and murine preadipocytes were treated with DTG either during differentiation into mature brown/beige adipocytes or postdifferentiation. Lipid accumulation, lipolysis, ß-adrenergic response, adipogenic markers, mitochondrial respiration, and insulin response were analyzed. RESULTS: Two-week administration of DTG to female mice reduced energy expenditure, which was accompanied by decreased uncoupling protein 1 (UCP1) expression in brown/beige adipose tissues. In vitro studies showed that DTG significantly reduced brown adipogenic markers, especially UCP1 in brown and beige adipocytes, whereas drugs from other classes did not. Furthermore, a loss of UCP1 by DTG led to a decrease in mitochondrial complex IV component, followed by a reduction in mitochondrial respiratory capacity and reduced insulin-stimulated glucose uptake. CONCLUSIONS: Our findings show that DTG targets UCP1 and mitochondrial functions in brown and beige adipocytes and disrupts thermogenic functions in preclinical models, providing the potential mechanisms by which DTG suppresses energy expenditure leading to weight gain.


Assuntos
Adipócitos Bege , Insulinas , Feminino , Humanos , Camundongos , Animais , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo , Adipócitos Bege/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Canais Iônicos/genética , Canais Iônicos/metabolismo , Termogênese/genética , Mitocôndrias/metabolismo , Metabolismo Energético/fisiologia , Aumento de Peso , Insulinas/metabolismo
9.
Physiol Rep ; 10(10): e15245, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35581741

RESUMO

Serotonin is an important mediator modulating behavior, metabolism, sleep, control of breathing, and upper airway function, but the role of aging in serotonin-mediated effects has not been previously defined. Our study aimed to examine the effect of brain serotonin deficiency on breathing during sleep and metabolism in younger and older mice. We measured breathing during sleep, hypercapnic ventilatory response (HCVR), CO2 production (VCO2 ), and O2 consumption (VO2 ) in 16-18-week old and 40-44-week old mice with deficiency of tryptophan hydroxylase 2 (Tph2), which regulates serotonin synthesis specifically in neurons, compared to Tph2+/+ mice. As expected, aging decreased VCO2 and VO2 . Tph2 knockout resulted in an increase in both metabolic indexes and no interaction between age and the genotype was observed. During wakefulness, neither age nor genotype had an effect on minute ventilation. The genotype did not affect hypercapnic sensitivity in younger mice. During sleep, Tph2-/- mice showed significant decreases in maximal inspiratory flow in NREM sleep, respiratory rate, and oxyhemoglobin saturation in REM sleep, compared to wildtype, regardless of age. Neither serotonin deficiency nor aging affected the frequency of flow limited breaths (a marker of upper airway closure) or apneas. Serotonin deficiency increased the amount and efficiency of sleep only in older animals. In conclusion, younger Tph2-/- mice were able to defend their ventilation and phenotypically did not differ from wildtype during wakefulness. In contrast, both young and old Tph2-/- mice showed sleep-related hypoventilation, which was manifested by hypoxemia during REM sleep.


Assuntos
Respiração , Serotonina , Animais , Encéfalo/metabolismo , Hipercapnia , Camundongos , Serotonina/metabolismo , Sono REM/fisiologia
10.
JCI Insight ; 6(9)2021 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-33784258

RESUMO

Perilipin 2 (PLIN2) is a lipid droplet (LD) protein in ß cells that increases under nutritional stress. Downregulation of PLIN2 is often sufficient to reduce LD accumulation. To determine whether PLIN2 positively or negatively affects ß cell function under nutritional stress, PLIN2 was downregulated in mouse ß cells, INS1 cells, and human islet cells. ß Cell-specific deletion of PLIN2 in mice on a high-fat diet reduced glucose-stimulated insulin secretion (GSIS) in vivo and in vitro. Downregulation of PLIN2 in INS1 cells blunted GSIS after 24-hour incubation with 0.2 mM palmitic acid. Downregulation of PLIN2 in human pseudoislets cultured at 5.6 mM glucose impaired both phases of GSIS, indicating that PLIN2 is critical for GSIS. Downregulation of PLIN2 decreased specific OXPHOS proteins in all 3 models and reduced oxygen consumption rates in INS1 cells and mouse islets. Moreover, we found that PLIN2-deficient INS1 cells increased the distribution of a fluorescent oleic acid analog to mitochondria and showed signs of mitochondrial stress, as indicated by susceptibility to fragmentation and alterations of acyl-carnitines and glucose metabolites. Collectively, PLIN2 in ß cells has an important role in preserving insulin secretion, ß cell metabolism, and mitochondrial function under nutritional stress.


Assuntos
Secreção de Insulina/genética , Células Secretoras de Insulina/metabolismo , Gotículas Lipídicas/metabolismo , Perilipina-2/genética , Estresse Fisiológico/genética , Animais , Carnitina/análogos & derivados , Carnitina/metabolismo , Dieta Hiperlipídica , Regulação para Baixo , Glucose/metabolismo , Humanos , Técnicas In Vitro , Ilhotas Pancreáticas , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Ácido Oleico/metabolismo , Fosforilação Oxidativa , Estresse Oxidativo/genética , Consumo de Oxigênio/genética , Perilipina-2/metabolismo , Ratos
11.
Sleep ; 44(6)2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-33624805

RESUMO

STUDY OBJECTIVES: Obesity leads to obstructive sleep apnea (OSA), which is recurrent upper airway obstruction during sleep, and obesity hypoventilation syndrome (OHS), hypoventilation during sleep resulting in daytime hypercapnia. Impaired leptin signaling in the brain was implicated in both conditions, but mechanisms are unknown. We have previously shown that leptin stimulates breathing and treats OSA and OHS in leptin-deficient ob/ob mice and leptin-resistant diet-induced obese mice and that leptin's respiratory effects may occur in the dorsomedial hypothalamus (DMH). We hypothesized that leptin receptor LepRb-deficient db/db mice have obesity hypoventilation and that restoration of leptin signaling in the DMH will increase ventilation during sleep in these animals. METHODS: We measured arterial blood gas in unanesthetized awake db/db mice. We subsequently infected these animals with Ad-LepRb or control Ad-mCherry virus into the DMH and measured ventilation during sleep as well as CO2 production after intracerebroventricular (ICV) infusions of phosphate-buffered saline or leptin. RESULTS: Awake db/db mice had elevated CO2 levels in the arterial blood. Ad-LepRb infection resulted in LepRb expression in the DMH neurons in a similar fashion to wildtype mice. In LepRb-DMH db/db mice, ICV leptin shortened REM sleep and increased inspiratory flow, tidal volume, and minute ventilation during NREM sleep without any effect on the quality of NREM sleep or CO2 production. Leptin had no effect on upper airway obstruction in these animals. CONCLUSION: Leptin stimulates breathing and treats obesity hypoventilation acting on LepRb-positive neurons in the DMH.


Assuntos
Leptina , Receptores para Leptina , Animais , Hipotálamo/metabolismo , Leptina/metabolismo , Camundongos , Camundongos Obesos , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Sono
12.
Ann Neurol ; 88(3): 513-525, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32557841

RESUMO

OBJECTIVE: To examine associations of molecular markers of brain insulin signaling with Alzheimer disease (AD) and cognition among older persons with or without diabetes. METHODS: This clinical-pathologic study was derived from a community-based cohort study, the Religious Orders Study. We studied 150 individuals (mean age at death =87 years, 48% women): 75 with and 75 without diabetes (matched by sex on age at death and education). Using enzyme-linked immunosorbent assay, immunohistochemistry, and ex vivo stimulation of brain tissue with insulin, we assessed insulin signaling in the postmortem middle frontal gyrus cortex. Postmortem data documented AD neuropathology. Clinical evaluations documented cognitive function proximate to death, based on 17 neuropsychological tests. In adjusted regression analyses, we examined associations of brain insulin signaling with diabetes, AD, and level of cognition. RESULTS: Brain insulin receptor substrate-1 (IRS1) phosphorylation (pS307 IRS1/total IRS1) and serine/threonine-protein kinase (AKT) phosphorylation (pT308 AKT1/total AKT1) were similar in persons with or without diabetes. AKT phosphorylation was associated with the global AD pathology score (p = 0.001). In contrast, IRS1 phosphorylation was not associated with AD (p = 0.536). No other associations of insulin signaling were found with the global AD score, including when using the ex vivo brain insulin stimulation method. In secondary analyses, normalized pT308 AKT1 was positively correlated with both the amyloid burden and tau tangle density, and no other associations of brain insulin signaling with neuropathology were observed. Moreover, normalized pT308 AKT1 was associated with a lower level of global cognitive function (estimate = -0.212, standard error = 0.097; p = 0.031). INTERPRETATION: Brain AKT phosphorylation, a critical node in the signaling of insulin and other growth factors, is associated with AD neuropathology and lower cognitive function. ANN NEUROL 2020;88:513-525.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Cognição/fisiologia , Insulina/metabolismo , Idoso de 80 Anos ou mais , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Feminino , Humanos , Masculino , Transdução de Sinais
13.
Obes Surg ; 29(12): 3941-3947, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31290107

RESUMO

BACKGROUND: Bariatric surgery is associated with improved cardiovascular outcomes and also affects lipid levels, but few studies have compared the effects of Roux-en-Y gastric bypass (RYGB) surgery with those of laparoscopic sleeve gastrectomy (LSG) on serum fatty acid levels. The present study compares the effects of RYGB and LSG surgeries on serum fatty acid levels. METHODS: The study participants were women who were undergoing either RYGB or LSG and body mass index (BMI)-matched controls. Fasting blood samples to measure glucose, insulin, and fatty acids were drawn at baseline and at 6 and 18 months from baseline. RESULTS: Serum fatty acid data were available for 57 participants at baseline, of whom 56 had data at 6 months and 41 had data at 18 months from baseline. Compared with baseline, serum non-esterified fatty acids (NEFAs) levels were significantly higher at 6 and 18 months in the LSG group compared with the RYGB group. In the RYGB group, 2 saturated fatty acids (SFAs), 2 monounsaturated fatty acids (MUFAs), and 1 polyunsaturated fatty acid (PUFA) were significantly decreased after surgery, compared with those of the LSG group. CONCLUSIONS: A significant increase in NEFAs was seen after LSG, compared with RYGB. Compared with the LSG group, several serum fatty acids were significantly reduced after RYGB. TRIAL REGISTRATION: NCT01228097.


Assuntos
Ácidos Graxos/sangue , Gastrectomia , Derivação Gástrica , Obesidade Mórbida/cirurgia , Adulto , Biomarcadores/sangue , Estudos de Casos e Controles , Feminino , Seguimentos , Gastrectomia/métodos , Humanos , Pessoa de Meia-Idade , Obesidade Mórbida/sangue , Período Pós-Operatório , Resultado do Tratamento
14.
Am J Respir Crit Care Med ; 199(6): 773-783, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30309268

RESUMO

RATIONALE: Leptin treats upper airway obstruction and alveolar hypoventilation in leptin-deficient ob/ob mice. However, obese humans and mice with diet-induced obesity (DIO) are resistant to leptin because of poor permeability of the blood-brain barrier. We propose that intranasal leptin will bypass leptin resistance and treat sleep-disordered breathing in obesity. OBJECTIVES: To assess if intranasal leptin can treat obesity hypoventilation and upper airway obstruction during sleep in mice with DIO. METHODS: Male C57BL/6J mice were fed with a high-fat diet for 16 weeks. A single dose of leptin (0.4 mg/kg) or BSA (vehicle) were administered intranasally or intraperitoneally, followed by either sleep studies (n = 10) or energy expenditure measurements (n = 10). A subset of mice was treated with leptin daily for 14 days for metabolic outcomes (n = 20). In a separate experiment, retrograde viral tracers were used to examine connections between leptin receptors and respiratory motoneurons. MEASUREMENTS AND MAIN RESULTS: Acute intranasal, but not intraperitoneal, leptin decreased the number of oxygen desaturation events in REM sleep, and increased ventilation in non-REM and REM sleep, independently of metabolic effects. Chronic intranasal leptin decreased food intake and body weight, whereas intraperitoneal leptin had no effect. Intranasal leptin induced signal transducer and activator of transcription 3 phosphorylation in hypothalamic and medullary centers, whereas intraperitoneal leptin had no effect. Leptin receptor-positive cells were synaptically connected to respiratory motoneurons. CONCLUSIONS: In mice with DIO, intranasal leptin bypassed leptin resistance and significantly attenuated sleep-disordered breathing independently of body weight.


Assuntos
Leptina/metabolismo , Absorção Nasal/fisiologia , Obesidade/complicações , Síndromes da Apneia do Sono/complicações , Síndromes da Apneia do Sono/fisiopatologia , Sono/fisiologia , Animais , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais
15.
Cell Rep ; 14(10): 2413-25, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26947067

RESUMO

p53 is well known for its tumor suppressor role, but this protein also has a poorly understood role in the regulation of metabolism. Human studies have implicated a common polymorphism at codon 72 of p53 in diabetic and pre-diabetic phenotypes. To understand this role, we utilized a humanized mouse model of the p53 codon 72 variants and monitored these mice following challenge with a high-fat diet (HFD). Mice with the arginine 72 (R72) variant of p53 developed more-severe obesity and glucose intolerance on a HFD, compared to mice with the proline 72 variant (P72). R72 mice developed insulin resistance, islet hypertrophy, increased infiltration of immune cells, and fatty liver disease. Gene expression analyses and studies with small-molecule inhibitors indicate that the p53 target genes Tnf and Npc1l1 underlie this phenotype. These results shed light on the role of p53 in obesity, metabolism, and inflammation.


Assuntos
Predisposição Genética para Doença , Obesidade/genética , Polimorfismo de Nucleotídeo Único , Proteína Supressora de Tumor p53/genética , Animais , Peso Corporal/genética , Dieta Hiperlipídica , Teste de Tolerância a Glucose , Humanos , Resistência à Insulina , Fígado/metabolismo , Fígado/patologia , Masculino , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Obesidade/fisiopatologia , Pâncreas/metabolismo , Pâncreas/patologia , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/metabolismo
16.
Am J Physiol Regul Integr Comp Physiol ; 309(8): R835-44, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26269523

RESUMO

IL-15Rα is the widely expressed primary binding partner for IL-15. Because of the wide distribution in nonlymphoid tissues like skeletal muscle, adipose, or liver, IL-15/IL-15Rα take part in physiological and metabolic processes not directly related to immunity. In fast muscle, lack of IL-15Rα promotes an oxidative switch, with increased mitochondrial biogenesis and fatigue resistance. These effects are predicted to reproduce some of the benefits of exercise and, therefore, improve energy homeostasis. However, the direct effects of IL-15Rα on metabolism and obesity are currently unknown. We report that mice lacking IL-15Rα (IL-15Rα(-/-)) are resistant to diet-induced obesity (DIO). High-fat diet-fed IL-15Rα(-/-) mice have less body and liver fat accumulation than controls. The leaner phenotype is associated with increased energy expenditure and enhanced fatty acid oxidation by muscle mitochondria. Despite being protected against DIO, IL-15Rα(-/-) are hyperglycemic and insulin-resistant. These findings identify novel roles for IL-15Rα in metabolism and obesity.


Assuntos
Metabolismo Energético/fisiologia , Regulação da Expressão Gênica/fisiologia , Subunidade alfa de Receptor de Interleucina-15/metabolismo , Interleucina-15/metabolismo , Músculo Esquelético/metabolismo , Obesidade/metabolismo , Animais , Glicemia , Composição Corporal , Temperatura Corporal , Ácidos Graxos/metabolismo , Teste de Tolerância a Glucose , Homeostase , Insulina/metabolismo , Interleucina-15/genética , Subunidade alfa de Receptor de Interleucina-15/genética , Camundongos , Camundongos Knockout , Obesidade/genética , Termografia
17.
Methods Mol Biol ; 1150: 273-81, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24744006

RESUMO

Reversal of terminally differentiated somatic cells to ground-state pluripotency has rejuvenated our hopes of generating patient-specific stem cells for therapeutic use in regenerative medicine and drug screening. Originally generated using defined exogenous protein-coding DNA, several methods have been described in reprogramming somatic cells into iPSC. Majority of published methods seek to improve or refine the techniques of reprogramming. This chapter describes reprogramming to pluripotency using miRNAs.


Assuntos
Reprogramação Celular/genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , MicroRNAs/genética , Animais , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Células HEK293 , Humanos , Lentivirus/genética , Lentivirus/fisiologia , Camundongos
18.
J Cell Sci ; 125(Pt 18): 4179-87, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23077173

RESUMO

The ability to generate pluripotent stem cells from a variety of cell and tissue sources through the ectopic expression of a specific set of transcription factors has revolutionized regenerative biology. The development of this reprogramming technology not only makes it possible to perform basic research on human stem cells that do not have to be derived from embryos, but also allows patient-specific cells and tissues to be generated for therapeutic use. Optimizing this process will probably lead to a better and more efficient means of generating pluripotent stem cells. Here, we discuss recent findings that show that, in addition to transcription factors, microRNAs can promote pluripotent reprogramming and can even substitute for these pluripotency transcription factors in some cases. Taking into consideration that microRNAs have the potential to be used as small-molecule therapeutics, such findings open new possibilities for both pluripotent stem cell reprogramming and the reprogramming of cells into other cell lineages.


Assuntos
Reprogramação Celular/genética , MicroRNAs/metabolismo , Células-Tronco Pluripotentes/metabolismo , Animais , Sequência de Bases , Proliferação de Células , Humanos , MicroRNAs/genética , Dados de Sequência Molecular , Células-Tronco Pluripotentes/citologia , Fatores de Transcrição/metabolismo
19.
Cell Stem Cell ; 8(4): 376-88, 2011 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-21474102

RESUMO

Transcription factor-based cellular reprogramming has opened the way to converting somatic cells to a pluripotent state, but has faced limitations resulting from the requirement for transcription factors and the relative inefficiency of the process. We show here that expression of the miR302/367 cluster rapidly and efficiently reprograms mouse and human somatic cells to an iPSC state without a requirement for exogenous transcription factors. This miRNA-based reprogramming approach is two orders of magnitude more efficient than standard Oct4/Sox2/Klf4/Myc-mediated methods. Mouse and human miR302/367 iPSCs display similar characteristics to Oct4/Sox2/Klf4/Myc-iPSCs, including pluripotency marker expression, teratoma formation, and, for mouse cells, chimera contribution and germline contribution. We found that miR367 expression is required for miR302/367-mediated reprogramming and activates Oct4 gene expression, and that suppression of Hdac2 is also required. Thus, our data show that miRNA and Hdac-mediated pathways can cooperate in a powerful way to reprogram somatic cells to pluripotency.


Assuntos
Reprogramação Celular/genética , MicroRNAs/fisiologia , Células-Tronco Pluripotentes/citologia , Animais , Humanos , Fator 4 Semelhante a Kruppel , Camundongos , Fatores de Transcrição/fisiologia
20.
J Mol Biol ; 379(2): 201-11, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18448117

RESUMO

Gene regulation during development is an important biological activity that leads to synthesis of biomolecules at specific locations and specific times. The single tropomyosin gene of Caenorhabditis elegans, tmy-1/lev-11, produces four isoforms of protein: two from the external promoter and two from the internal promoter. We investigated the internal promoter of tropomyosin to identify sequences that regulate expression of tmy-1 in the pharynx and intestine. By promoter deletion of tmy-1 reporters as well as by database analyses, a 100-bp fragment that contained binding sequences for a GATA factor, for a chicken CdxA homolog, and for a forkhead factor was identified. Both the forkhead and CdxA binding sequences contributed to pharyngeal and intestinal expression. In addition, the GATA site also influenced intestinal expression of tmy-1 reporter. We showed that ELT-2 and PHA-4 proteins interact directly with the GATA and forkhead binding sequences, respectively, in gel mobility shift assays. RNA interference knockdown of elt-2 diminished tmy-1::gfp expression in the intestine. In contrast to RNA interference knockdown of pha-4, expression of tmy-1::gfp in pha-4;smg-1 mutants was slightly weaker than that of the wild type. Ectopic expression of PHA-4 and ELT-2 by heat shock was sufficient to elicit widespread expression of tmy-1::lacZ reporter in embryos. We found no indication of a synergistic relation between ELT-2 and PHA-4. Based on our data, PHA-4 and CdxA function as general transcription factors for pharyngeal and intestinal regulation of tmy-1. We present models by which ELT-2, PHA-4, and CdxA orchestrate expression from the internal promoter of tmy-1.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans , Fatores de Transcrição GATA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Intestinos/fisiologia , Faringe/fisiologia , Transativadores/metabolismo , Tropomiosina , Animais , Animais Geneticamente Modificados , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Sequência de Bases , Caenorhabditis elegans/anatomia & histologia , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/genética , Linhagem da Célula , Galinhas , Fatores de Transcrição GATA/genética , Genes Reporter , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Intestinos/anatomia & histologia , Dados de Sequência Molecular , Faringe/anatomia & histologia , Regiões Promotoras Genéticas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferência de RNA , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transativadores/genética , Tropomiosina/genética , Tropomiosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...