Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Kidney Int ; 102(3): 560-576, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35654224

RESUMO

Acute kidney injury is a frequent complication in the clinical setting and associated with significant morbidity and mortality. Preconditioning with short-term caloric restriction is highly protective against kidney injury in rodent ischemia reperfusion injury models. However, the underlying mechanisms are unknown hampering clinical translation. Here, we examined the molecular basis of caloric restriction-mediated protection to elucidate the principles of kidney stress resistance. Analysis of an RNAseq dataset after caloric restriction identified Cyp4a12a, a cytochrome exclusively expressed in male mice, to be strongly downregulated after caloric restriction. Kidney ischemia reperfusion injury robustly induced acute kidney injury in male mice and this damage could be markedly attenuated by pretreatment with caloric restriction. In females, damage was significantly less pronounced and preconditioning with caloric restriction had only little effect. Tissue concentrations of the metabolic product of Cyp4a12a, 20-hydroxyeicosatetraenoic acid (20-HETE), were found to be significantly reduced by caloric restriction. Conversely, intraperitoneal supplementation of 20-HETE in preconditioned males partly abrogated the protective potential of caloric restriction. Interestingly, this effect was accompanied by a partial reversal of caloric restriction--induced changes in protein but not RNA expression pointing towards inflammation, endoplasmic reticulum stress and lipid metabolism. Thus, our findings provide an insight into the mechanisms underlying kidney protection by caloric restriction. Hence, understanding the mediators of preconditioning is an important prerequisite for moving towards translation to the clinical setting.


Assuntos
Injúria Renal Aguda , Traumatismo por Reperfusão , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/prevenção & controle , Animais , Restrição Calórica , Ácidos Hidroxieicosatetraenoicos/metabolismo , Ácidos Hidroxieicosatetraenoicos/farmacologia , Rim/metabolismo , Masculino , Camundongos , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle
2.
J Endocrinol ; 251(3): 195-206, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34612845

RESUMO

Vascular reactivity of adipose tissue (AT) is hypothesized to play an important role in the development of obesity. However, the exact role of vascular reactivity in the development of obesity remains unclear. In this study, we investigated the chronological changes in vascular reactivity and the microenvironments of the visceral AT (VAT) and subcutaneous AT (SAT) in lean and obese mice. Changes in blood flow levels induced by a ß-adrenoceptor agonist (isoproterenol) were significantly lower in the VAT of the mice fed a high-fat diet (HFD) for 1 and 12 weeks than those in the VAT of the mice fed a low-fat diet (LFD) for the same period; no significant change was observed in the SAT of any mouse group, suggesting depot-specific vascular reactivity of AT. Moreover, the hypoxic area and the expression of genes associated with angiogenesis and macrophage recruitment were increased in the VAT (but not in the SAT) of mice fed an HFD for 1 week compared with mice fed an LFD. These changes occurred with no morphological changes, including those related to adipocyte size, AT vessel density, and the diameter and pericyte coverage of the endothelium, suggesting a determinant role of vascular reactivity in the type of AT remodeling. The suppression of vascular reactivity was accompanied by increased endothelin1 (Edn1) gene expression and extracellular matrix (ECM) stiffness only in the VAT, implying enhanced contractile activities of the vasculature and ECM. The results suggest a depot-specific role of vascular reactivity in AT remodeling during the development of obesity.


Assuntos
Gordura Intra-Abdominal/irrigação sanguínea , Neovascularização Patológica , Obesidade/induzido quimicamente , Animais , Dieta Hiperlipídica , Masculino , Camundongos , Camundongos Obesos , Obesidade/patologia
3.
Cell Rep ; 27(11): 3182-3198.e9, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31189104

RESUMO

Variations in the human FTO gene have been linked to obesity and altered connectivity of the dopaminergic neurocircuitry. Here, we report that fat mass and obesity-associated protein (FTO) in dopamine D2 receptor-expressing medium spiny neurons (D2 MSNs) of mice regulate the excitability of these cells and control their striatopallidal globus pallidus external (GPe) projections. Lack of FTO in D2 MSNs translates into increased locomotor activity to novelty, associated with altered timing behavior, without impairing the ability to control actions or affecting reward-driven and conditioned behavior. Pharmacological manipulations of dopamine D1 receptor (D1R)- or D2R-dependent pathways in these animals reveal altered responses to D1- and D2-MSN-mediated control of motor output. These findings reveal a critical role for FTO to control D2 MSN excitability, their projections to the GPe, and behavioral responses to novelty.


Assuntos
Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Neurônios Dopaminérgicos/metabolismo , Comportamento Exploratório , Locomoção , Potenciais de Ação , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Animais , Neurônios Dopaminérgicos/fisiologia , Feminino , Globo Pálido/citologia , Globo Pálido/metabolismo , Globo Pálido/fisiologia , Masculino , Camundongos , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Recompensa
4.
Genes Brain Behav ; 17(8): e12516, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30133126

RESUMO

Adhesion molecules of the immunoglobulin superfamily (IgSF) are essential for neuronal synapse development across evolution and control various aspects of synapse formation and maturation. Neph2, also known as Kirrel3, is an IgSF adhesion molecule implicated in synapse formation, synaptic transmission and ultrastructure. In humans, defects in the NEPH2 gene have been associated with neurodevelopmental disorders such as Jacobsen syndrome, intellectual disability, and autism-spectrum disorders. However, the precise role in development and function of the nervous system is still unclear. Here, we present the histomorphological and phenotypical analysis of a constitutive Neph2-knockout mouse line. Knockout mice display defects in auditory sensory processing, motor skills, and hyperactivity in the home-cage analysis. Olfactory, memory and metabolic testing did not differ from controls. Despite the wide-spread expression of Neph2 in various brain areas, no gross anatomic defects could be observed. Neph2 protein could be located at the cerebellar pinceaux. It interacted with the pinceau core component neurofascin and other synaptic proteins thus suggesting a possible role in cerebellar synapse formation and circuit assembly. Our results suggest that Neph2/Kirrel3 acts on the synaptic ultrastructural level and neuronal wiring rather than on ontogenetic events affecting macroscopic structure. Neph2-knockout mice may provide a valuable rodent model for research on autism spectrum diseases and neurodevelopmental disorders.


Assuntos
Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Animais , Proteínas de Transporte/genética , Adesão Celular/fisiologia , Imunoglobulinas/fisiologia , Camundongos , Camundongos Knockout , Neurogênese , Neurônios/metabolismo , Sinapses/metabolismo
5.
Br J Nutr ; 120(7): 763-776, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30109842

RESUMO

Intra-uterine growth restriction (IUGR) is associated with adverse metabolic outcome later in life. Healthy mice challenged with a Western-style diet (WSD) accumulated less body fat when previously fed a diet containing large lipid globules (complex lipid matrix (CLM)). This study was designed to clarify whether an early-life CLM diet mitigates 'programmed' visceral adiposity and associated metabolic sequelae after IUGR. In rats, IUGR was induced either by bilateral uterine vessel ligation (LIG) or sham operation (i.e. intra-uterine stress) of the dam on gestational day 19. Offspring from non-operated (NOP) dams served as controls. Male offspring of all groups were either fed CLM or 'normal matrix' control diet (CTRL) from postnatal days (PND) 15 to 42. Thereafter, animals were challenged with a mild WSD until dissection (PND 98). Fat mass (micro computer-tomograph scan; weight of fat compartments), circulating metabolic markers and expression of 'metabolic' genes (quantitative real-time PCR) were assessed. CLM diet significantly reduced visceral fat mass in LIG at PND 40. At dissection, visceral fat mass, fasted blood glucose, TAG and leptin concentrations were significantly increased in LIG-CTRL v. NOP-CTRL, and significantly decreased in LIG-CLM v. LIG-CTRL. Gene expression levels of leptin (mesenteric fat) and insulin-like growth factor 1 (liver) were significantly reduced in LIG-CLM v. LIG-CTRL. In conclusion, early-life CLM diet mitigated the adverse metabolic phenotype after utero-placental insufficiency. The supramolecular structure of dietary lipids may be a novel aspect of nutrient quality that has to be considered in the context of primary prevention of obesity and metabolic disease in at-risk populations.


Assuntos
Glicemia/metabolismo , Dieta , Gorduras na Dieta/farmacologia , Retardo do Crescimento Fetal/metabolismo , Fenômenos Fisiológicos da Nutrição do Lactente , Gordura Intra-Abdominal/metabolismo , Lipídeos/farmacologia , Animais , Biomarcadores/metabolismo , Dieta Ocidental , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/metabolismo , Feminino , Humanos , Lactente , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Leptina/sangue , Ligadura , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipídeos/administração & dosagem , Lipídeos/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Mesentério , Gravidez , Ratos Wistar , Triglicerídeos/sangue , Útero/cirurgia
6.
Cell Metab ; 26(1): 198-211.e5, 2017 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-28683287

RESUMO

Olfactory inputs help coordinate food appreciation and selection, but their role in systemic physiology and energy balance is poorly understood. Here we demonstrate that mice upon conditional ablation of mature olfactory sensory neurons (OSNs) are resistant to diet-induced obesity accompanied by increased thermogenesis in brown and inguinal fat depots. Acute loss of smell perception after obesity onset not only abrogated further weight gain but also improved fat mass and insulin resistance. Reduced olfactory input stimulates sympathetic nerve activity, resulting in activation of ß-adrenergic receptors on white and brown adipocytes to promote lipolysis. Conversely, conditional ablation of the IGF1 receptor in OSNs enhances olfactory performance in mice and leads to increased adiposity and insulin resistance. These findings unravel a new bidirectional function for the olfactory system in controlling energy homeostasis in response to sensory and hormonal signals.


Assuntos
Obesidade/metabolismo , Obesidade/fisiopatologia , Neurônios Receptores Olfatórios/metabolismo , Olfato , Termogênese , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Marrom/fisiopatologia , Animais , Dieta Hiperlipídica/efeitos adversos , Metabolismo Energético , Resistência à Insulina , Fator de Crescimento Insulin-Like I/metabolismo , Lipólise , Camundongos , Obesidade/etiologia , Neurônios Receptores Olfatórios/patologia , Receptores Adrenérgicos beta/metabolismo , Aumento de Peso
7.
Nat Cell Biol ; 18(3): 328-36, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26900752

RESUMO

Activation of brown adipose tissue (BAT) controls energy homeostasis in rodents and humans and has emerged as an innovative strategy for the treatment of obesity and type 2 diabetes mellitus. Here we show that ageing- and obesity-associated dysfunction of brown fat coincides with global microRNA downregulation due to reduced expression of the microRNA-processing node Dicer1. Consequently, heterozygosity of Dicer1 in BAT aggravated diet-induced-obesity (DIO)-evoked deterioration of glucose metabolism. Analyses of differential microRNA expression during preadipocyte commitment and mouse models of progeria, longevity and DIO identified miR-328 as a regulator of BAT differentiation. Reducing miR-328 blocked preadipocyte commitment, whereas miR-328 overexpression instigated BAT differentiation and impaired muscle progenitor commitment-partly through silencing of the ß-secretase Bace1. Loss of Bace1 enhanced brown preadipocyte specification in vitro and was overexpressed in BAT of obese and progeroid mice. In vivo Bace1 inhibition delayed DIO-induced weight gain and improved glucose tolerance and insulin sensitivity. These experiments reveal Dicer1-miR-328-Bace1 signalling as a determinant of BAT function, and highlight the potential of Bace1 inhibition as a therapeutic approach to improve not only neurodegenerative diseases but also ageing- and obesity-associated impairments of BAT function.


Assuntos
Tecido Adiposo Marrom/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Ácido Aspártico Endopeptidases/genética , Diferenciação Celular/fisiologia , RNA Helicases DEAD-box/genética , MicroRNAs/genética , Ribonuclease III/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , RNA Helicases DEAD-box/metabolismo , Metabolismo Energético/fisiologia , Homeostase/fisiologia , Resistência à Insulina/fisiologia , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Obesidade/genética , Obesidade/metabolismo , Ribonuclease III/metabolismo
8.
Cell Rep ; 9(4): 1495-506, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25456138

RESUMO

Activation of c-Jun N-terminal kinase 1 (JNK1)- and inhibitor of nuclear factor kappa-B kinase 2 (IKK2)-dependent signaling plays a crucial role in the development of obesity-associated insulin and leptin resistance not only in peripheral tissues but also in the CNS. Here, we demonstrate that constitutive JNK activation in agouti-related peptide (AgRP)-expressing neurons of the hypothalamus is sufficient to induce weight gain and adiposity in mice as a consequence of hyperphagia. JNK activation increases spontaneous action potential firing of AgRP cells and causes both neuronal and systemic leptin resistance. Similarly, activation of IKK2 signaling in AgRP neurons also increases firing of these cells but fails to cause obesity and leptin resistance. In contrast to JNK activation, IKK2 activation blunts insulin signaling in AgRP neurons and impairs systemic glucose homeostasis. Collectively, these experiments reveal both overlapping and nonredundant effects of JNK- and IKK-dependent signaling in AgRP neurons, which cooperate in the manifestation of the metabolic syndrome.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Quinase I-kappa B/metabolismo , Resistência à Insulina , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neurônios/enzimologia , Obesidade/enzimologia , Potenciais de Ação/efeitos dos fármacos , Adiposidade/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Glucose/metabolismo , Homeostase/efeitos dos fármacos , Insulina/metabolismo , Leptina/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Proteínas Mutantes/metabolismo , Neurônios/efeitos dos fármacos
9.
Cell Metab ; 20(4): 678-86, 2014 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-25295788

RESUMO

Ceramides increase during obesity and promote insulin resistance. Ceramides vary in acyl-chain lengths from C14:0 to C30:0 and are synthesized by six ceramide synthase enzymes (CerS1-6). It remains unresolved whether obesity-associated alterations of specific CerSs and their defined acyl-chain length ceramides contribute to the manifestation of metabolic diseases. Here we reveal that CERS6 mRNA expression and C16:0 ceramides are elevated in adipose tissue of obese humans, and increased CERS6 expression correlates with insulin resistance. Conversely, CerS6-deficient (CerS6(Δ/Δ)) mice exhibit reduced C16:0 ceramides and are protected from high-fat-diet-induced obesity and glucose intolerance. CerS6 deletion increases energy expenditure and improves glucose tolerance, not only in CerS6(Δ/Δ) mice, but also in brown adipose tissue- (CerS6(ΔBAT)) and liver-specific (CerS6(ΔLIVER)) CerS6 knockout mice. CerS6 deficiency increases lipid utilization in BAT and liver. These experiments highlight CerS6 inhibition as a specific approach for the treatment of obesity and type 2 diabetes mellitus, circumventing the side effects of global ceramide synthesis inhibition.


Assuntos
Ceramidas/metabolismo , Intolerância à Glucose , Esfingosina N-Aciltransferase/metabolismo , Tecido Adiposo Marrom/metabolismo , Animais , Índice de Massa Corporal , Dieta Hiperlipídica , Feminino , Humanos , Peroxidação de Lipídeos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Obesidade/metabolismo , Obesidade/patologia , PPAR gama/genética , PPAR gama/metabolismo , Esfingosina N-Aciltransferase/deficiência , Esfingosina N-Aciltransferase/genética , Aumento de Peso
10.
Nat Immunol ; 15(5): 423-30, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24681566

RESUMO

Obesity and resistance to insulin are closely associated with the development of low-grade inflammation. Interleukin 6 (IL-6) is linked to obesity-associated inflammation; however, its role in this context remains controversial. Here we found that mice with an inactivated gene encoding the IL-6Rα chain of the receptor for IL-6 in myeloid cells (Il6ra(Δmyel) mice) developed exaggerated deterioration of glucose homeostasis during diet-induced obesity, due to enhanced resistance to insulin. Tissues targeted by insulin showed increased inflammation and a shift in macrophage polarization. IL-6 induced expression of the receptor for IL-4 and augmented the response to IL-4 in macrophages in a cell-autonomous manner. Il6ra(Δmyel) mice were resistant to IL-4-mediated alternative polarization of macrophages and exhibited enhanced susceptibility to lipopolysaccharide (LPS)-induced endotoxemia. Our results identify signaling via IL-6 as an important determinant of the alternative activation of macrophages and assign an unexpected homeostatic role to IL-6 in limiting inflammation.


Assuntos
Endotoxemia/imunologia , Resistência à Insulina , Interleucina-6/metabolismo , Ativação de Macrófagos , Macrófagos/imunologia , Obesidade/imunologia , Animais , Células Cultivadas , Humanos , Resistência à Insulina/genética , Resistência à Insulina/imunologia , Interleucina-4/imunologia , Interleucina-6/genética , Lipopolissacarídeos/imunologia , Ativação de Macrófagos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Receptores de Interleucina-6/genética , Transdução de Sinais/genética
11.
Nat Commun ; 5: 3427, 2014 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-24619096

RESUMO

Dysregulation of hypothalamic-pituitary-adrenal (HPA) axis activity leads to debilitating neuroendocrine or metabolic disorders such as Cushing's syndrome (CS). Glucocorticoids control HPA axis activity through negative feedback to the pituitary gland and the central nervous system (CNS). However, the cellular mechanisms involved are poorly understood, particularly in the CNS. Here we show that, in mice, selective loss of TrkB signalling in cholecystokinin (CCK)-GABAergic neurons induces glucocorticoid resistance, resulting in increased corticotrophin-releasing hormone expression, chronic hypercortisolism, adrenocortical hyperplasia, glucose intolerance and mature-onset obesity, reminiscent of the human CS phenotype. Interestingly, obesity is not due to hyperphagia or decreased energy expenditure, but is associated with increased de novo lipogenesis in the liver. Our study therefore identifies CCK neurons as a novel and critical cellular component of the HPA axis, and demonstrates the requirement of TrkB for the transmission of glucocorticoid signalling.


Assuntos
Colecistocinina/metabolismo , Síndrome de Cushing/metabolismo , Neurônios GABAérgicos/metabolismo , Glicoproteínas de Membrana/metabolismo , Obesidade/metabolismo , Proteínas Tirosina Quinases/metabolismo , Animais , Composição Corporal/efeitos dos fármacos , Calorimetria Indireta , Colecistocinina/genética , Síndrome de Cushing/genética , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Neurônios GABAérgicos/efeitos dos fármacos , Immunoblotting , Hibridização In Situ , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Mifepristona/farmacologia , Obesidade/genética , Proteínas Tirosina Quinases/genética
12.
Cell Metab ; 18(3): 445-55, 2013 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-24011078

RESUMO

Brown adipose tissue (BAT) is a critical regulator of glucose, lipid, and energy homeostasis, and its activity is tightly controlled by the sympathetic nervous system. However, the mechanisms underlying CNS-dependent control of BAT sympathetic nerve activity (SNA) are only partly understood. Here, we demonstrate that catecholaminergic neurons in the locus coeruleus (LC) adapt their firing frequency to extracellular glucose concentrations in a K(ATP)-channel-dependent manner. Inhibiting K(ATP)-channel-dependent control of neuronal activity via the expression of a variant K(ATP) channel in tyrosine-hydroxylase-expressing neurons and in neurons of the LC enhances diet-induced obesity in mice. Obesity results from decreased energy expenditure, lower steady-state BAT SNA, and an attenuated ability of centrally applied glucose to activate BAT SNA. This impairs the thermogenic transcriptional program of BAT. Collectively, our data reveal a role of K(ATP)-channel-dependent neuronal excitability in catecholaminergic neurons in maintaining thermogenic BAT sympathetic tone and energy homeostasis.


Assuntos
Tecido Adiposo Marrom/metabolismo , Neurônios Colinérgicos/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Sistema Nervoso Simpático/metabolismo , Animais , Neurônios Colinérgicos/efeitos dos fármacos , Dieta Hiperlipídica , Metabolismo Energético/efeitos dos fármacos , Glucose/farmacologia , Locus Cerúleo/efeitos dos fármacos , Locus Cerúleo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Obesidade/etiologia , Obesidade/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Tirosina 3-Mono-Oxigenase/metabolismo
13.
Nat Neurosci ; 16(8): 1042-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23817550

RESUMO

Dopaminergic (DA) signaling governs the control of complex behaviors, and its deregulation has been implicated in a wide range of diseases. Here we demonstrate that inactivation of the Fto gene, encoding a nucleic acid demethylase, impairs dopamine receptor type 2 (D2R) and type 3 (D3R) (collectively, 'D2-like receptor')-dependent control of neuronal activity and behavioral responses. Conventional and DA neuron-specific Fto knockout mice show attenuated activation of G protein-coupled inwardly-rectifying potassium (GIRK) channel conductance by cocaine and quinpirole. Impaired D2-like receptor-mediated autoinhibition results in attenuated quinpirole-mediated reduction of locomotion and an enhanced sensitivity to the locomotor- and reward-stimulatory actions of cocaine. Analysis of global N(6)-methyladenosine (m(6)A) modification of mRNAs using methylated RNA immunoprecipitation coupled with next-generation sequencing in the midbrain and striatum of Fto-deficient mice revealed increased adenosine methylation in a subset of mRNAs important for neuronal signaling, including many in the DA signaling pathway. Several proteins encoded by these mRNAs had altered expression levels. Collectively, FTO regulates the demethylation of specific mRNAs in vivo, and this activity relates to the control of DA transmission.


Assuntos
Dopamina/fisiologia , Neurônios Dopaminérgicos/enzimologia , Mesencéfalo/fisiologia , Oxigenases de Função Mista/fisiologia , Oxo-Ácido-Liases/fisiologia , Adenina/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Cocaína/farmacologia , Corpo Estriado/fisiologia , Neurônios Dopaminérgicos/fisiologia , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Metilação , Metiltransferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxigenases de Função Mista/deficiência , Oxigenases de Função Mista/genética , Oxo-Ácido-Liases/deficiência , Oxo-Ácido-Liases/genética , Fenótipo , Quimpirol/farmacologia , Processamento Pós-Transcricional do RNA , RNA Mensageiro/metabolismo , Receptores de Dopamina D2/deficiência , Receptores de Dopamina D2/fisiologia , Receptores de Dopamina D3/fisiologia , Recompensa , Transdução de Sinais/efeitos dos fármacos
14.
J Mol Med (Berl) ; 91(6): 739-48, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23344677

RESUMO

MicroRNAs have emerged as essential regulators of gene expression and may play important roles in a variety of human disorders. To understand the role of microRNA-mediated gene regulation in the kidney, we deleted the microRNA-processing enzyme Dicer in developing renal tubules and parts of the ureteric bud in mice. Genetic deletion of Dicer resulted in renal failure and death of the animals at 4-6 weeks of age. Interestingly, the kidneys of microRNA-deficient animals were small due to a reduced number of nephrons and showed massive hydronephrosis due to ureteropelvic junction obstruction. This phenotype is reminiscent of congenital anomalies of the kidney and urinary tract (CAKUT), an important group of human disorders characterized by a combination of renal hypoplasia with congenital abnormalities of the urinary tract. We used metanephric kidney cultures to examine the developmental defects underlying these pathologies. Dicer knockout kidneys showed a significant reduction of tubular branching explaining renal hypoplasia. Moreover, the ureters of these kidneys showed an altered morphology and impaired motility. These functional changes went along with altered expression of smooth muscle actin implying a defect in the differentiation of ureteric smooth muscle cells. In addition, we show the polycystic kidney disease gene Pkd1 to be a target of miR-20 implying that this interaction may contribute to the molecular basis for the cystogenesis in our model. In conclusion, these data demonstrate an essential role for microRNA-dependent gene regulation in mammalian kidney development and suggest that deregulation of microRNAs may underlie CAKUT, the most important group of renal disorders in humans.


Assuntos
MicroRNAs/genética , Refluxo Vesicoureteral/genética , Animais , Caderinas/genética , RNA Helicases DEAD-box/genética , Feminino , Células HEK293 , Humanos , Integrases/genética , Rim/anormalidades , Masculino , Camundongos , Camundongos Knockout , Ribonuclease III/genética , Canais de Cátion TRPP/genética , Sistema Urinário/anormalidades , Anormalidades Urogenitais
15.
PLoS One ; 8(1): e54247, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23349837

RESUMO

Obesity and associated metabolic disturbances, such as increased circulating fatty acids cause prolonged low grade activation of inflammatory signaling pathways in liver, skeletal muscle, adipose tissue and even in the CNS. Activation of inflammatory pathways in turn impairs insulin signaling, ultimately leading to obesity-associated type 2 diabetes mellitus. Conventional JNK-1 knock out mice are protected from high fat diet-induced insulin resistance, characterizing JNK-1-inhibition as a potential approach to improve glucose metabolism in obese patients. However, the cell type-specific role of elevated JNK-1 signaling as present during the course of obesity has not been fully elucidated yet. To investigate the functional contribution of altered JNK-1 activation in skeletal muscle, we have generated a ROSA26 insertion mouse strain allowing for Cre-activatable expression of a JNK-1 constitutive active construct (JNK(C)). To examine the consequence of skeletal muscle-restricted JNK-1 overactivation in the development of insulin resistance and glucose metabolism, JNK(C) mice were crossed to Mck-Cre mice yielding JNK(SM-C) mice. However, despite increased muscle-specific JNK activation, energy homeostasis and glucose metabolism in JNK(SM-C) mice remained largely unaltered compared to controls. In line with these findings, obese mice with skeletal muscle specific disruption of JNK-1, did not affect energy and glucose homeostasis. These experiments indicate that JNK-1 activation in skeletal muscle does not account for the major effects on diet-induced, JNK-1-mediated deterioration of insulin action and points towards a so far underappreciated role of JNK-1 in other tissues than skeletal muscle during the development of obesity-associated insulin resistance.


Assuntos
Glucose/metabolismo , Homeostase , Resistência à Insulina , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Músculo Esquelético/metabolismo , Obesidade/metabolismo , Animais , Western Blotting , Composição Corporal , Dieta Hiperlipídica/efeitos adversos , Metabolismo Energético , Feminino , Expressão Gênica , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteína Quinase 8 Ativada por Mitógeno/genética , Obesidade/etiologia , Obesidade/genética , Fosforilação , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , RNA não Traduzido , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
PLoS Genet ; 8(11): e1003021, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23144624

RESUMO

Fusion and fission of mitochondria maintain the functional integrity of mitochondria and protect against neurodegeneration, but how mitochondrial dysfunctions trigger neuronal loss remains ill-defined. Prohibitins form large ring complexes in the inner membrane that are composed of PHB1 and PHB2 subunits and are thought to function as membrane scaffolds. In Caenorhabditis elegans, prohibitin genes affect aging by moderating fat metabolism and energy production. Knockdown experiments in mammalian cells link the function of prohibitins to membrane fusion, as they were found to stabilize the dynamin-like GTPase OPA1 (optic atrophy 1), which mediates mitochondrial inner membrane fusion and cristae morphogenesis. Mutations in OPA1 are associated with dominant optic atrophy characterized by the progressive loss of retinal ganglion cells, highlighting the importance of OPA1 function in neurons. Here, we show that neuron-specific inactivation of Phb2 in the mouse forebrain causes extensive neurodegeneration associated with behavioral impairments and cognitive deficiencies. We observe early onset tau hyperphosphorylation and filament formation in the hippocampus, demonstrating a direct link between mitochondrial defects and tau pathology. Loss of PHB2 impairs the stability of OPA1, affects mitochondrial ultrastructure, and induces the perinuclear clustering of mitochondria in hippocampal neurons. A destabilization of the mitochondrial genome and respiratory deficiencies manifest in aged neurons only, while the appearance of mitochondrial morphology defects correlates with tau hyperphosphorylation in the absence of PHB2. These results establish an essential role of prohibitin complexes for neuronal survival in vivo and demonstrate that OPA1 stability, mitochondrial fusion, and the maintenance of the mitochondrial genome in neurons depend on these scaffolding proteins. Moreover, our findings establish prohibitin-deficient mice as a novel genetic model for tau pathologies caused by a dysfunction of mitochondria and raise the possibility that tau pathologies are associated with other neurodegenerative disorders caused by deficiencies in mitochondrial dynamics.


Assuntos
Mitocôndrias , Doenças Neurodegenerativas , Neurônios , Atrofia Óptica Autossômica Dominante , Proteínas Repressoras , Animais , Apoptose , Proteínas de Caenorhabditis elegans , Genoma Mitocondrial , Fusão de Membrana , Camundongos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Membranas Mitocondriais/metabolismo , Morfogênese , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo , Neurônios/patologia , Atrofia Óptica Autossômica Dominante/genética , Atrofia Óptica Autossômica Dominante/metabolismo , Fosforilação , Proibitinas , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
17.
Cell Metab ; 13(6): 720-8, 2011 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-21641553

RESUMO

Dopaminergic midbrain neurons integrate signals on food palatability and food-associated reward into the complex control of energy homeostasis. To define the role of insulin receptor (IR) signaling in this circuitry, we inactivated IR signaling in tyrosine hydroxylase (Th)-expressing cells of mice (IR(ΔTh)). IR inactivation in Th-expressing cells of mice resulted in increased body weight, increased fat mass, and hyperphagia. While insulin acutely stimulated firing frequency in 50% of dopaminergic VTA/SN neurons, this response was abolished in IR(ΔTh) mice. Moreover, these mice exhibited an altered response to cocaine under food-restricted conditions. Taken together, these data provide in vivo evidence for a critical role of insulin signaling in catecholaminergic neurons to control food intake and energy homeostasis.


Assuntos
Metabolismo Energético/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Insulina/farmacologia , Neurônios/metabolismo , Potenciais de Ação , Adiposidade , Animais , Calorimetria Indireta , Catecolaminas/metabolismo , Cocaína/farmacologia , Ingestão de Alimentos/genética , Expressão Gênica , Hiperinsulinismo/genética , Mesencéfalo/citologia , Mesencéfalo/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Neurônios/efeitos dos fármacos , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Transdução de Sinais , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
18.
Nat Cell Biol ; 13(4): 434-46, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21441927

RESUMO

The contribution of altered post-transcriptional gene silencing to the development of insulin resistance and type 2 diabetes mellitus so far remains elusive. Here, we demonstrate that expression of microRNA (miR)-143 and 145 is upregulated in the liver of genetic and dietary mouse models of obesity. Induced transgenic overexpression of miR-143, but not miR-145, impairs insulin-stimulated AKT activation and glucose homeostasis. Conversely, mice deficient for the miR-143-145 cluster are protected from the development of obesity-associated insulin resistance. Quantitative-mass-spectrometry-based analysis of hepatic protein expression in miR-143-overexpressing mice revealed miR-143-dependent downregulation of oxysterol-binding-protein-related protein (ORP) 8. Reduced ORP8 expression in cultured liver cells impairs the ability of insulin to induce AKT activation, revealing an ORP8-dependent mechanism of AKT regulation. Our experiments provide direct evidence that dysregulated post-transcriptional gene silencing contributes to the development of obesity-induced insulin resistance, and characterize the miR-143-ORP8 pathway as a potential target for the treatment of obesity-associated diabetes.


Assuntos
Glucose/metabolismo , Insulina/metabolismo , MicroRNAs/metabolismo , Obesidade/genética , Obesidade/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Dieta , Ativação Enzimática , Resistência à Insulina , Fígado/enzimologia , Camundongos , Camundongos Obesos , Camundongos Transgênicos , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética
19.
Cell Metab ; 12(3): 237-49, 2010 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-20816090

RESUMO

The contribution of interleukin (IL)-6 signaling in obesity-induced inflammation remains controversial. To specifically define the role of hepatic IL-6 signaling in insulin action and resistance, we have generated mice with hepatocyte-specific IL-6 receptor (IL-6R) alpha deficiency (IL-6Ralpha(L-KO) mice). These animals showed no alterations in body weight and fat content but exhibited a reduction in insulin sensitivity and glucose tolerance. Impaired glucose metabolism originated from attenuated insulin-stimulated glucose transport in skeletal muscle and fat. Surprisingly, hepatic IL-6Ralpha-disruption caused an exaggerated inflammatory response during euglycemic hyperinsulinemic clamp analysis, as revealed by increased expression of IL-6, TNF-alpha, and IL-10, as well as enhanced activation of inflammatory signaling such as phosphorylation of IkappaBalpha. Neutralization of TNF-alpha or ablation of Kupffer cells restored glucose tolerance in IL-6Ralpha(L-KO) mice. Thus, our results reveal an unexpected role for hepatic IL-6 signaling to limit hepatic inflammation and to protect from local and systemic insulin resistance.


Assuntos
Inflamação/metabolismo , Insulina/metabolismo , Interleucina-6/metabolismo , Fígado/citologia , Fígado/patologia , Transdução de Sinais/fisiologia , Adiposidade , Animais , Metabolismo Energético , Glucose/metabolismo , Glicogênio/biossíntese , Homeostase , Humanos , Resistência à Insulina/fisiologia , Interleucina-10/metabolismo , Células de Kupffer/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Knockout , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
20.
Proc Natl Acad Sci U S A ; 107(33): 14875-80, 2010 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-20679202

RESUMO

The neuronal circuits involved in the regulation of feeding behavior and energy expenditure are soft-wired, reflecting the relative activity of the postsynaptic neuronal system, including the anorexigenic proopiomelanocortin (POMC)-expressing cells of the arcuate nucleus. We analyzed the synaptic input organization of the melanocortin system in lean rats that were vulnerable (DIO) or resistant (DR) to diet-induced obesity. We found a distinct difference in the quantitative and qualitative synaptology of POMC cells between DIO and DR animals, with a significantly greater number of inhibitory inputs in the POMC neurons in DIO rats compared with DR rats. When exposed to a high-fat diet (HFD), the POMC cells of DIO animals lost synapses, whereas those of DR rats recruited connections. In both DIO rats and mice, the HFD-triggered loss of synapses on POMC neurons was associated with increased glial ensheathment of the POMC perikarya. The altered synaptic organization of HFD-fed animals promoted increased POMC tone and a decrease in the stimulatory connections onto the neighboring neuropeptide Y (NPY) cells. Exposure to HFD was associated with reactive gliosis, and this affected the structure of the blood-brain barrier such that the POMC and NPY cell bodies and dendrites became less accessible to blood vessels. Taken together, these data suggest that consumption of an HFD has a major impact on the cytoarchitecture of the arcuate nucleus in vulnerable subjects, with changes that might be irreversible due to reactive gliosis.


Assuntos
Dieta , Gliose/metabolismo , Melanocortinas/metabolismo , Obesidade/metabolismo , Sinapses/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Núcleo Arqueado do Hipotálamo/fisiopatologia , Gorduras na Dieta/efeitos adversos , Feminino , Gliose/etiologia , Hipotálamo/metabolismo , Hipotálamo/patologia , Hipotálamo/fisiopatologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica , Neurônios/metabolismo , Neurônios/ultraestrutura , Neuropeptídeo Y/metabolismo , Obesidade/etiologia , Pró-Opiomelanocortina/metabolismo , Ratos , Ratos Sprague-Dawley , Transmissão Sináptica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA