Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Exp Hematol ; 129: 104124, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37898316

RESUMO

Erythroid terminal differentiation and maturation depend on an enormous energy supply. During periods of fasting, ketone bodies from the liver are transported into circulation and utilized as crucial fuel for peripheral tissues. However, the effects of fasting or ketogenesis on erythroid behavior remain unknown. Here, we generated a mouse model with insufficient ketogenesis by conditionally knocking out the gene encoding the hepatocyte-specific ketogenic enzyme hydroxymethylglutary-CoA synthase 2 (Hmgcs2 KO). Intriguingly, erythroid maturation was enhanced with boosted fatty acid synthesis in the bone marrow of a hepatic Hmgcs2 KO mouse under fasting conditions, suggesting that systemic ketogenesis has a profound effect on erythropoiesis. Moreover, we observed significantly activated fatty acid synthesis and mevalonate pathways along with reduced histone acetylation in immature erythrocytes under a less systemic ketogenesis condition. Our findings revealed a new insight into erythroid differentiation, in which metabolic homeostasis and histone acetylation mediated by ketone bodies are essential factors in adaptation toward nutrient deprivation and stressed erythropoiesis.


Assuntos
Histonas , Hidroximetilglutaril-CoA Sintase , Camundongos , Animais , Histonas/metabolismo , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Corpos Cetônicos/genética , Corpos Cetônicos/metabolismo , Fígado/metabolismo , Jejum/fisiologia , Ácidos Graxos/metabolismo
2.
Mol Metab ; 61: 101494, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35421611

RESUMO

OBJECTIVE: Aberrant ketogenesis is correlated with the degree of steatosis in non-alcoholic fatty liver disease (NAFLD) patients, and an inborn error of ketogenesis (mitochondrial HMG-CoA synthase deficiency) is commonly associated with the development of the fatty liver. Here we aimed to determine the impact of Hmgcs2-mediated ketogenesis and its modulations on the development and treatment of fatty liver disease. METHODS: Loss- and gain-of-ketogenic function models, achieved by Hmgcs2 knockout and overexpression, respectively, were utilized to investigate the role of ketogenesis in the hepatic lipid accumulation during postnatal development and in a high-fat diet-induced NAFLD mouse model. RESULTS: Ketogenic function was decreased in NAFLD mice with a reduction in Hmgcs2 expression. Mice lacking Hmgcs2 developed spontaneous fatty liver phenotype during postnatal development, which was rescued by a shift to a low-fat dietary composition via early weaning. Hmgcs2 heterozygous adult mice, which exhibited lower ketogenic activity, were more susceptible to diet-induced NAFLD development, whereas HMGCS2 overexpression in NAFLD mice improved hepatosteatosis and glucose homeostasis. CONCLUSIONS: Our study adds new knowledge to the field of ketone body metabolism and shows that Hmgcs2-mediated ketogenesis modulates hepatic lipid regulation under a fat-enriched nutritional environment. The regulation of hepatic ketogenesis may be a viable therapeutic strategy in the prevention and treatment of hepatosteatosis.


Assuntos
Dieta Hiperlipídica , Hidroximetilglutaril-CoA Sintase , Cetose , Hepatopatia Gordurosa não Alcoólica , Animais , Dieta Hiperlipídica/efeitos adversos , Humanos , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Corpos Cetônicos/genética , Corpos Cetônicos/metabolismo , Cetose/genética , Cetose/metabolismo , Lipídeos , Camundongos , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo
3.
Sci Rep ; 11(1): 16359, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34381166

RESUMO

A better understanding of the secondary injury mechanisms that occur after traumatic spinal cord injury (SCI) is essential for the development of novel neuroprotective strategies linked to the restoration of metabolic deficits. We and others have shown that Ketogenic diet (KD), a high fat, moderate in proteins and low in carbohydrates is neuroprotective and improves behavioural outcomes in rats with acute SCI. Ketones are alternative fuels for mitochondrial ATP generation, and can modulate signaling pathways via targeting specific receptors. Here, we demonstrate that ad libitum administration of KD for 7 days after SCI rescued mitochondrial respiratory capacity, increased parameters of mitochondrial biogenesis, affected the regulation of mitochondrial-related genes, and activated the NRF2-dependent antioxidant pathway. This study demonstrates that KD improves post-SCI metabolism by rescuing mitochondrial function and supports the potential of KD for treatment of acute SCI in humans.


Assuntos
Medula Cervical/patologia , Metabolismo Energético/genética , Expressão Gênica/genética , Genes Mitocondriais/genética , Mitocôndrias/genética , Traumatismos da Medula Espinal/genética , Animais , Dieta Cetogênica/métodos , Modelos Animais de Doenças , Corpos Cetônicos/genética , Masculino , Biogênese de Organelas , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/genética , Transdução de Sinais/genética , Medula Espinal/patologia , Traumatismos da Medula Espinal/patologia
4.
J Neurochem ; 157(6): 1930-1945, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33539571

RESUMO

Ketogenic diets (KDs) alter brain metabolism. Multiple mechanisms may account for their effects, and different brain regions may variably respond. Here, we considered how a KD affects brain neuron and astrocyte transcription. We placed male C57Bl6/N mice on either a 3-month KD or chow diet, generated enriched neuron and astrocyte fractions, and used RNA-Seq to assess transcription. Neurons from KD-treated mice generally showed transcriptional pathway activation while their astrocytes showed a mix of transcriptional pathway suppression and activation. The KD especially affected pathways implicated in mitochondrial and endoplasmic reticulum function, insulin signaling, and inflammation. An unbiased analysis of KD-associated expression changes strongly implicated transcriptional pathways altered in AD, which prompted us to explore in more detail the potential molecular relevance of a KD to AD. Our results indicate a KD differently affects neurons and astrocytes, and provide unbiased evidence that KD-induced brain effects are potentially relevant to neurodegenerative diseases such as AD.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , Dieta Cetogênica/métodos , Corpos Cetônicos/metabolismo , Neurônios/metabolismo , Transcrição Gênica/fisiologia , Animais , Dieta Cetogênica/tendências , Corpos Cetônicos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL
5.
Biochimie ; 183: 55-62, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33596448

RESUMO

Succinyl-CoA:3-oxoacid coenzyme A transferase deficiency (SCOTD) is a rare autosomal recessive disorder of ketone body utilization caused by mutations in OXCT1. We performed a systematic literature search and evaluated clinical, biochemical and genetic data on 34 previously published and 10 novel patients with SCOTD. Structural mapping and in silico analysis of protein variants is also presented. All patients presented with severe ketoacidotic episodes. Age at first symptoms ranged from 36 h to 3 years (median 7 months). About 70% of patients manifested in the first year of life, approximately one quarter already within the neonatal period. Two patients died, while the remainder (95%) were alive at the time of the report. Almost all the surviving patients (92%) showed normal psychomotor development and no neurologic abnormalities. A total of 29 missense mutations are reported. Analysis of the published crystal structure of the human SCOT enzyme, paired with both sequence-based and structure-based methods to predict variant pathogenicity, provides insight into the biochemical consequences of the reported variants. Pathogenic variants cluster in SCOT protein regions that affect certain structures of the protein. The described pathogenic variants can be viewed in an interactive map of the SCOT protein at https://michelanglo.sgc.ox.ac.uk/r/oxct. This comprehensive data analysis provides a systematic overview of all cases of SCOTD published to date. Although SCOTD is a rather benign disorder with often favourable outcome, metabolic crises can be life-threatening or even fatal. As the diagnosis can only be made by enzyme studies or mutation analyses, SCOTD may be underdiagnosed.


Assuntos
Acidose , Encefalopatias Metabólicas Congênitas , Coenzima A-Transferases/deficiência , Mutação de Sentido Incorreto , Transtornos do Neurodesenvolvimento , Acidose/enzimologia , Acidose/genética , Encefalopatias Metabólicas Congênitas/enzimologia , Encefalopatias Metabólicas Congênitas/genética , Coenzima A-Transferases/química , Coenzima A-Transferases/genética , Coenzima A-Transferases/metabolismo , Cristalografia por Raios X , Humanos , Corpos Cetônicos/química , Corpos Cetônicos/genética , Corpos Cetônicos/metabolismo , Transtornos do Neurodesenvolvimento/enzimologia , Transtornos do Neurodesenvolvimento/genética , Domínios Proteicos
6.
Signal Transduct Target Ther ; 6(1): 54, 2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33558457

RESUMO

In addition to their use in relieving the symptoms of various diseases, ketogenic diets (KDs) have also been adopted by healthy individuals to prevent being overweight. Herein, we reported that prolonged KD exposure induced cardiac fibrosis. In rats, KD or frequent deep fasting decreased mitochondrial biogenesis, reduced cell respiration, and increased cardiomyocyte apoptosis and cardiac fibrosis. Mechanistically, increased levels of the ketone body ß-hydroxybutyrate (ß-OHB), an HDAC2 inhibitor, promoted histone acetylation of the Sirt7 promoter and activated Sirt7 transcription. This in turn inhibited the transcription of mitochondrial ribosome-encoding genes and mitochondrial biogenesis, leading to cardiomyocyte apoptosis and cardiac fibrosis. Exogenous ß-OHB administration mimicked the effects of a KD in rats. Notably, increased ß-OHB levels and SIRT7 expression, decreased mitochondrial biogenesis, and increased cardiac fibrosis were detected in human atrial fibrillation heart tissues. Our results highlighted the unknown detrimental effects of KDs and provided insights into strategies for preventing cardiac fibrosis in patients for whom KDs are medically necessary.


Assuntos
Dieta Cetogênica/efeitos adversos , Fibrose/genética , Corpos Cetônicos/genética , Sirtuínas/genética , Ácido 3-Hidroxibutírico/metabolismo , Ácido 3-Hidroxibutírico/farmacologia , Acetilação , Animais , Apoptose/genética , Modelos Animais de Doenças , Jejum/metabolismo , Fibrose/etiologia , Fibrose/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Histona Desacetilase 2/antagonistas & inibidores , Histona Desacetilase 2/genética , Histonas/genética , Histonas/metabolismo , Humanos , Corpos Cetônicos/metabolismo , Masculino , Miócitos Cardíacos/metabolismo , Biogênese de Organelas , Ratos
7.
Eur J Pharmacol ; 893: 173804, 2021 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-33347826

RESUMO

There is no known single therapeutic drug for treating hypercholesterolemia that comes with negligible systemic side effects. In the current study, using next generation RNA sequencing approach in mouse embryonic fibroblasts we discovered that two structurally related flavonoid compounds. Apigenin and Chrysin exhibited moderate blocking ability of multiple transcripts that regulate rate limiting enzymes in the cholesterol biosynthesis pathway. The observed decrease in cholesterol biosynthesis pathway correlated well with an increase in transcripts involved in generation and trafficking of ketone bodies as evident by the upregulation of Bdh1 and Slc16a6 transcripts. The hypocholesterolemic potential of Apigenin and Chrysin at higher concentrations along with their ability to generate ketogenic substrate especially during embryonic stage is useful or detrimental for embryonic health is not clear and still debatable. Our study will serve as a steppingstone to further the investigation in whole animal studies and also in translating this knowledge to human studies.


Assuntos
Anticolesterolemiantes/farmacologia , Apigenina/farmacologia , Colesterol/biossíntese , Fibroblastos/efeitos dos fármacos , Flavonoides/farmacologia , Perfilação da Expressão Gênica , Corpos Cetônicos/metabolismo , Lipogênese/efeitos dos fármacos , Transcriptoma , Animais , Anticolesterolemiantes/química , Apigenina/química , Células Cultivadas , Fibroblastos/metabolismo , Flavonoides/química , Regulação da Expressão Gênica , Corpos Cetônicos/genética , Lipogênese/genética , Camundongos , Estrutura Molecular
8.
Nutrients ; 12(3)2020 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-32192146

RESUMO

Ketone bodies (KBs), comprising ß-hydroxybutyrate, acetoacetate and acetone, are a set of fuel molecules serving as an alternative energy source to glucose. KBs are mainly produced by the liver from fatty acids during periods of fasting, and prolonged or intense physical activity. In diabetes, mainly type-1, ketoacidosis is the pathological response to glucose malabsorption. Endogenous production of ketone bodies is promoted by consumption of a ketogenic diet (KD), a diet virtually devoid of carbohydrates. Despite its recently widespread use, the systemic impact of KD is only partially understood, and ranges from physiologically beneficial outcomes in particular circumstances to potentially harmful effects. Here, we firstly review ketone body metabolism and molecular signaling, to then link the understanding of ketone bodies' biochemistry to controversies regarding their putative or proven medical benefits. We overview the physiological consequences of ketone bodies' consumption, focusing on (i) KB-induced histone post-translational modifications, particularly ß-hydroxybutyrylation and acetylation, which appears to be the core epigenetic mechanisms of activity of ß-hydroxybutyrate to modulate inflammation; (ii) inflammatory responses to a KD; (iii) proven benefits of the KD in the context of neuronal disease and cancer; and (iv) consequences of the KD's application on cardiovascular health and on physical performance.


Assuntos
Diabetes Mellitus Tipo 1 , Dieta Cetogênica , Epigênese Genética , Neoplasias , Doenças do Sistema Nervoso , Ácido 3-Hidroxibutírico/metabolismo , Acetoacetatos/metabolismo , Animais , Diabetes Mellitus Tipo 1/dietoterapia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Epigenômica , Humanos , Corpos Cetônicos/genética , Corpos Cetônicos/metabolismo , Cetose/dietoterapia , Cetose/genética , Cetose/metabolismo , Cetose/patologia , Metabolômica , Neoplasias/dietoterapia , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Doenças do Sistema Nervoso/dietoterapia , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/patologia
9.
Curr Alzheimer Res ; 15(10): 917-927, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29852870

RESUMO

BACKGROUND: Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by a multi-factorial etiology that is not completely understood. Donepezil is a first-line acetylcholinesterase inhibitor used for the treatment of AD that has been found, in addition to its potent acetylcholinesterase inhibitory effect, to act through other non-cholinergic mechanisms such as affecting mitochondrial biogenesis through peroxisome proliferator-activated receptor gamma coactivator (PGC1α). Mitochondrial biogenesis and PGC-1α, at least in part, are associated with hepatic fatty acid oxidation and ketogenesis. Whether donepezil regulates ketogenesis in AD treatment remains unclear. Ketogenesis is important in the progression of AD and is a critical consideration during the therapeutic strategy selection for AD. Thus, our goals were to determine the differences in ketone bodies in patients with AD who were taking donepezil treatment and those who were not, to elucidate the potential effect of AD and donepezil therapy on ketone body metabolic parameters, and to discover the effect of donepezil therapy on ketogenesis in patients with AD. METHODS: Cross-sectional analysis was performed on plasma collected from 145 individuals, namely, elderly adults as healthy controls (n=30), newly diagnosed patients with AD (n=30), patients with AD who responded to donepezil therapy (n=48) and patients with AD who did not respond to donepezil therapy (n=37). Gas chromatography-mass spectrometry was performed to quantify the lipids in the plasma. The level of ß-hydroxybutyrate, a metabolite, was determined by liquid chromatographytandem mass spectrometry, and to gain further insight into the effect of donepezil on ketogenesis, the effects of donepezil were investigated in a mouse model. RESULTS: The level of ß-hydroxybutyrate decreased in AD patients, and donepezil elevated the plasma level of ß-hydroxybutyrate. Donepezil increased the plasma and liver levels of ß-hydroxybutyrate in mice as well as the hepatic expression of PGC-1α and the mitochondrial expression of HMG-CoA synthetase 2 (HMGCS2) in response to fasting, causing a subsequent increase in ketogenesis. CONCLUSIONS: Our study revealed that impaired ketogenesis is a metabolic feature of AD. Donepezil had effects on ketogenesis in mice and reversed the decrease in the level of ß-hydroxybutyrate found in patients with AD.


Assuntos
Ácido 3-Hidroxibutírico/sangue , Doença de Alzheimer/sangue , Doença de Alzheimer/tratamento farmacológico , Inibidores da Colinesterase/uso terapêutico , Donepezila/uso terapêutico , Idoso , Idoso de 80 Anos ou mais , Animais , Glicemia/metabolismo , Colesterol/sangue , Estudos Transversais , Jejum/sangue , Ácidos Graxos/sangue , Feminino , Cromatografia Gasosa-Espectrometria de Massas , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Corpos Cetônicos/genética , Corpos Cetônicos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Fatores de Tempo
10.
J Inherit Metab Dis ; 40(6): 845-852, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28695376

RESUMO

Succinyl-CoA:3-oxoacid CoA transferase (SCOT, gene symbol OXCT1) deficiency is an autosomal recessive disorder in ketone body utilization that results in severe recurrent ketoacidotic episodes in infancy, including neonatal periods. More than 30 patients with this disorder have been reported and to our knowledge, their heterozygous parents and siblings have had no apparent ketoacidotic episodes. Over 5 years (2008-2012), we investigated several patients that presented with severe ketoacidosis and identified a heterozygous OXCT1 mutation in four of these cases (Case1 p.R281C, Case2 p.T435N, Case3 p.W213*, Case4 c.493delG). To confirm their heterozygous state, we performed a multiplex ligation-dependent probe amplification analysis on the OXCT1 gene which excluded the presence of large deletions or insertions in another allele. A sequencing analysis of subcloned full-length SCOT cDNA showed that wild-type cDNA clones were present at reasonable rates to mutant cDNA clones. Over the following 2 years (2013-2014), we analyzed OXCT1 mutations in six more patients presenting with severe ketoacidosis (blood pH ≦7.25 and total ketone body ≧10 mmol/L) with non-specific urinary organic acid profiles. Of these, a heterozygous OXCT1 mutation was found in two cases (Case5 p.G391D, Case6 p.R281C). Moreover, transient expression analysis revealed R281C and T435N mutants to be temperature-sensitive. This characteristic may be important because most patients developed ketoacidosis during infections. Our data indicate that heterozygous carriers of OXCT1 mutations can develop severe ketoacidotic episodes in conjunction with ketogenic stresses.


Assuntos
Acidose/genética , Acidose/patologia , Acil Coenzima A/deficiência , Coenzima A-Transferases/deficiência , Cetose/genética , Cetose/patologia , Acil Coenzima A/genética , Criança , Pré-Escolar , Coenzima A-Transferases/genética , DNA Complementar/genética , Feminino , Heterozigoto , Humanos , Lactente , Corpos Cetônicos/genética , Masculino , Mutação/genética
11.
Cell Physiol Biochem ; 41(5): 1766-1776, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28365686

RESUMO

BACKGROUND/AIMS: Stress response is determined by the brain, and the brain is a sensitive target for stress. Our previous experiments have confirmed that once the stress response is beyond the tolerable limit of the brain, particularly that of the hippocampus, it will have deleterious effects on hippocampal structure and function; however, the metabolic mechanisms for this are not well understood. METHODS: Here, we used morris water maze, elisa and gas chromatography-time of flight/mass spectrometry to observe the changes in cognition, neuropathology and metabolomics in the hippocampus of APP/PS1 mice and wild-type (C57) mice caused by chronic unpredictable mild stress (CUMS), we also further explored the correlation between cognition and metabolomics. RESULTS: We found that 4 weeks of CUMS aggravated cognitive impairment and increased amyloid-ß deposition in APP/PS1 mice, but did not affect C57 mice. Under non-stress conditions, compared with C57 mice, there were 8 different metabolites in APP/PS1 mice. However, following CUMS, 3 different metabolites were changed compared with untreated C57 mice. Compared to APP/PS1 mice, there were 7 different metabolites in APP/PS1+CUMS mice. Among these alterations, 3-hydroxybutyric acid, valine, serine, beta-alanine and o-phosphorylethanolamine, which are involved in sphingolipid metabolism, synthesis and degradation of ketone bodies, and amino acid metabolism. CONCLUSION: The results indicate that APP/PS1 mice are more vulnerable to stress than C57 mice, and the metabolic mechanisms of stress-related cognitive impairment in APP/PS1 mice are related to multiple pathways and networks, including sphingolipid metabolism, synthesis and degradation of ketone bodies, and amino acid metabolism.


Assuntos
Precursor de Proteína beta-Amiloide , Disfunção Cognitiva , Hipocampo , Metaboloma , Estresse Psicológico , Aminoácidos/genética , Aminoácidos/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Doença Crônica , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Corpos Cetônicos/genética , Corpos Cetônicos/metabolismo , Camundongos , Camundongos Transgênicos , Esfingolipídeos/genética , Esfingolipídeos/metabolismo , Estresse Psicológico/complicações , Estresse Psicológico/genética , Estresse Psicológico/metabolismo , Estresse Psicológico/patologia
12.
Sci Rep ; 6: 34909, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27708432

RESUMO

Ketone bodies have been shown to transiently stimulate food intake and modify energy homeostasis regulatory systems following cerebral infusion for a moderate period of time (<6 hours). As ketone bodies are usually enhanced during episodes of fasting, this effect might correspond to a physiological regulation. In contrast, ketone bodies levels remain elevated for prolonged periods during obesity, and thus could play an important role in the development of this pathology. In order to understand this transition, ketone bodies were infused through a catheter inserted in the carotid to directly stimulate the brain for a period of 24 hours. Food ingested and blood circulating parameters involved in metabolic control as well as glucose homeostasis were determined. Results show that ketone bodies infusion for 24 hours increased food intake associated with a stimulation of hypothalamic orexigenic neuropeptides. Moreover, insulinemia was increased and caused a decrease in glucose production despite an increased resistance to insulin. The present study confirms that ketone bodies reaching the brain stimulates food intake. Moreover, we provide evidence that a prolonged hyperketonemia leads to a dysregulation of energy homeostasis control mechanisms. Finally, this study shows that brain exposure to ketone bodies alters insulin signaling and consequently glucose homeostasis.


Assuntos
Ingestão de Alimentos/fisiologia , Hipotálamo/metabolismo , Corpos Cetônicos/metabolismo , Ácido 3-Hidroxibutírico/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Artérias Carótidas , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/genética , Regulação da Expressão Gênica , Homeostase , Hipotálamo/efeitos dos fármacos , Hipotálamo/fisiopatologia , Infusões Intra-Arteriais , Resistência à Insulina , Corpos Cetônicos/genética , Corpos Cetônicos/farmacologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Camundongos Endogâmicos C57BL , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fosfoenolpiruvato Carboxiquinase (ATP)/metabolismo , Pró-Opiomelanocortina/metabolismo , Simportadores/metabolismo
13.
Biochem Biophys Res Commun ; 473(2): 654-61, 2016 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-27021680

RESUMO

Previous studies have shown that high-fat diet (HFD)-induced obesity increases the acetoacetyl-CoA synthetase (AACS) gene expression in lipogenic tissue. To investigate the effect of obesity on the AACS gene in other tissues, we examined the alteration of AACS mRNA levels in HFD-fed mice. In situ hybridization revealed that AACS was observed in several regions of the embryo, including the backbone region (especially in the somite), and in the epiphysis of the adult femur. AACS mRNA expression in the adult femur was higher in HFD-fed mice than in normal-diet fed mice, but this increase was not observed in high sucrose diet (HSD)-induced obese mice. In addition, HFD-specific increases were observed in the 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) and interleukin (IL)-6 genes. Moreover, we detected higher AACS mRNA expression in the differentiated osteoclast cells (RAW 264), and found that AACS mRNA expression was significantly up-regulated by IL-6 treatment only in osteoclasts. These results indicate the novel function of the ketone body in bone metabolism. Because the abnormal activation of osteoclasts by IL-6 induces bone resorption, our data suggest that AACS and ketone bodies are important factors in the relationship between obesity and osteoporosis.


Assuntos
Osso e Ossos/patologia , Dieta Hiperlipídica/efeitos adversos , Corpos Cetônicos/metabolismo , Obesidade/etiologia , Obesidade/metabolismo , Osteoclastos/patologia , Animais , Osso e Ossos/metabolismo , Linhagem Celular , Coenzima A Ligases/genética , Inflamação/etiologia , Inflamação/genética , Inflamação/metabolismo , Interleucina-6/genética , Corpos Cetônicos/genética , Masculino , Camundongos , Camundongos Obesos , Obesidade/genética , Osteoclastos/metabolismo , RNA Mensageiro/genética , Regulação para Cima
14.
Biochim Biophys Acta ; 1852(3): 473-81, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25533124

RESUMO

SLC25A13 (citrin or aspartate-glutamate carrier 2) is located in the mitochondrial membrane in the liver and its genetic deficiency causes adult-onset type II citrullinemia (CTLN2). CTLN2 is one of the urea cycle disorders characterized by sudden-onset hyperammonemia due to reduced argininosuccinate synthase activity. This disorder is frequently accompanied with hepatosteatosis in the absence of obesity and ethanol consumption. However, the precise mechanism of steatogenesis remains unclear. The expression of genes associated with fatty acid (FA) and triglyceride (TG) metabolism was examined using liver samples obtained from 16 CTLN2 patients and compared with 7 healthy individuals. Although expression of hepatic genes associated with lipogenesis and TG hydrolysis was not changed, the mRNAs encoding enzymes/proteins involved in FA oxidation (carnitine palmitoyl-CoA transferase 1α, medium- and very-long-chain acyl-CoA dehydrogenases, and acyl-CoA oxidase 1), very-low-density lipoprotein secretion (microsomal TG transfer protein), and FA transport (CD36 and FA-binding protein 1), were markedly suppressed in CTLN2 patients. Serum concentrations of ketone bodies were also decreased in these patients, suggesting reduced mitochondrial ß-oxidation activity. Consistent with these findings, the expression of peroxisome proliferator-activated receptor α (PPARα), a master regulator of hepatic lipid metabolism, was significantly down-regulated. Hepatic PPARα expression was inversely correlated with severity of steatosis and circulating ammonia and citrulline levels. Additionally, phosphorylation of c-Jun-N-terminal kinase was enhanced in CTLN2 livers, which was likely associated with lower hepatic PPARα. Collectively, down-regulation of PPARα is associated with steatogenesis in CTLN2 patients. These findings provide a novel link between urea cycle disorder, lipid metabolism, and PPARα.


Assuntos
Citrulinemia/metabolismo , Regulação para Baixo , Fígado Gorduroso/metabolismo , Metabolismo dos Lipídeos , Mitocôndrias Hepáticas/metabolismo , PPAR alfa/biossíntese , Adulto , Citrulinemia/complicações , Citrulinemia/genética , Citrulinemia/patologia , Ácidos Graxos/genética , Ácidos Graxos/metabolismo , Fígado Gorduroso/etiologia , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Feminino , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Corpos Cetônicos/genética , Corpos Cetônicos/metabolismo , Masculino , Pessoa de Meia-Idade , Mitocôndrias Hepáticas/genética , Mitocôndrias Hepáticas/patologia , Proteínas de Transporte da Membrana Mitocondrial , PPAR alfa/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Triglicerídeos/genética , Triglicerídeos/metabolismo
15.
J Biol Chem ; 288(27): 19739-49, 2013 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-23689508

RESUMO

Preservation of bioenergetic homeostasis during the transition from the carbohydrate-laden fetal diet to the high fat, low carbohydrate neonatal diet requires inductions of hepatic fatty acid oxidation, gluconeogenesis, and ketogenesis. Mice with loss-of-function mutation in the extrahepatic mitochondrial enzyme CoA transferase (succinyl-CoA:3-oxoacid CoA transferase, SCOT, encoded by nuclear Oxct1) cannot terminally oxidize ketone bodies and develop lethal hyperketonemic hypoglycemia within 48 h of birth. Here we use this model to demonstrate that loss of ketone body oxidation, an exclusively extrahepatic process, disrupts hepatic intermediary metabolic homeostasis after high fat mother's milk is ingested. Livers of SCOT-knock-out (SCOT-KO) neonates induce the expression of the genes encoding peroxisome proliferator-activated receptor γ co-activator-1a (PGC-1α), phosphoenolpyruvate carboxykinase (PEPCK), pyruvate carboxylase, and glucose-6-phosphatase, and the neonate's pools of gluconeogenic alanine and lactate are each diminished by 50%. NMR-based quantitative fate mapping of (13)C-labeled substrates revealed that livers of SCOT-KO newborn mice synthesize glucose from exogenously administered pyruvate. However, the contribution of exogenous pyruvate to the tricarboxylic acid cycle as acetyl-CoA is increased in SCOT-KO livers and is associated with diminished terminal oxidation of fatty acids. After mother's milk provokes hyperketonemia, livers of SCOT-KO mice diminish de novo hepatic ß-hydroxybutyrate synthesis by 90%. Disruption of ß-hydroxybutyrate production increases hepatic NAD(+)/NADH ratios 3-fold, oxidizing redox potential in liver but not skeletal muscle. Together, these results indicate that peripheral ketone body oxidation prevents hypoglycemia and supports hepatic metabolic homeostasis, which is critical for the maintenance of glycemia during the adaptation to birth.


Assuntos
Coenzima A-Transferases , Gluconeogênese , Glucose/biossíntese , Hipoglicemia/metabolismo , Corpos Cetônicos/metabolismo , Fígado/metabolismo , Ácido 3-Hidroxibutírico/biossíntese , Ácido 3-Hidroxibutírico/genética , Animais , Animais Recém-Nascidos , Ciclo do Ácido Cítrico/efeitos dos fármacos , Ciclo do Ácido Cítrico/genética , Feminino , Glucose/genética , Hipoglicemia/genética , Corpos Cetônicos/genética , Fígado/patologia , Camundongos , Camundongos Knockout , NAD/genética , NAD/metabolismo , Oxirredução , Parto , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Ácido Pirúvico/farmacologia , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição
16.
Int J Neuropsychopharmacol ; 16(2): 271-8, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22717047

RESUMO

It is believed that multiple genetic variants with small individual effects contribute to the risk of alcohol dependence. Such polygenic effects are difficult to detect in genome-wide association studies that test for association of the phenotype with each single nucleotide polymorphism (SNP) individually. To overcome this challenge, gene-set analysis (GSA) methods that jointly test for the effects of pre-defined groups of genes have been proposed. Rather than testing for association between the phenotype and individual SNPs, these analyses evaluate the global evidence of association with a set of related genes enabling the identification of cellular or molecular pathways or biological processes that play a role in development of the disease. It is hoped that by aggregating the evidence of association for all available SNPs in a group of related genes, these approaches will have enhanced power to detect genetic associations with complex traits. We performed GSA using data from a genome-wide study of 1165 alcohol-dependent cases and 1379 controls from the Study of Addiction: Genetics and Environment (SAGE), for all 200 pathways listed in the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. Results demonstrated a potential role of the 'synthesis and degradation of ketone bodies' pathway. Our results also support the potential involvement of the 'neuroactive ligand-receptor interaction' pathway, which has previously been implicated in addictive disorders. These findings demonstrate the utility of GSA in the study of complex disease, and suggest specific directions for further research into the genetic architecture of alcohol dependence.


Assuntos
Alcoolismo/genética , Predisposição Genética para Doença/genética , Corpos Cetônicos/genética , Polimorfismo de Nucleotídeo Único/genética , Células Receptoras Sensoriais , Transdução de Sinais/genética , Ciclo Celular/genética , Bases de Dados Genéticas/estatística & dados numéricos , Feminino , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Corpos Cetônicos/metabolismo , Ligantes , Desequilíbrio de Ligação , Masculino , Fenótipo , Análise de Componente Principal
17.
Artigo em Inglês | MEDLINE | ID: mdl-22748507

RESUMO

Ruminants absorb little glucose and rely on hepatic gluconeogenesis and ketogenesis in the fed state to convert short-chain fatty acids produced during digestion into glucose and ketone bodies, respectively. In contrast to the non-ruminant response, fluxes through gluconeogenic and ketogenic pathways decrease during food restriction. Transcriptional regulation responsible for these unique food restriction responses has not been established. To determine the hepatic transcriptional response of ruminants to an acute drop in dietary nutrient supply, 102 yearling heifers were assigned to either ad libitum feeding or 24 h of food withdrawal in a randomized block design. Liver biopsies were obtained for microarray and quantitative real-time PCR analyses of gene expression. Plasma concentrations of non-esterified fatty acids were higher in food restricted heifers, while levels of ß-hydroxybutyrate, triacylglycerol, and glucose were decreased. Despite a decline in substrate supply and a lower hepatic production of glucose, expression of the key gluconeogenic enzymes pyruvate carboxylase, phosphoenolpyruvate carboxykinase and fructose-1,6-bisphosphatase was upregulated as in non-ruminants. Downregulation of cholesterolgenic genes and upregulation of fatty acid oxidative genes were consistent with SREBP-2 and PPARα control, respectively. Ketogenesis from short-chain fatty acids was downregulated, contrary to the non-ruminant response to food restriction. Short-chain fatty acids may exert transcriptional control in the ruminant liver similar to that demonstrated in the large intestine of non-ruminants.


Assuntos
Restrição Calórica , Perfilação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Corpos Cetônicos/biossíntese , Corpos Cetônicos/genética , Fígado/enzimologia , Ruminantes/genética , Ácido 3-Hidroxibutírico/sangue , Animais , Bovinos , Ácidos Graxos não Esterificados/sangue , Humanos , Redes e Vias Metabólicas/genética , Metaboloma/genética , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reprodutibilidade dos Testes , Ruminantes/sangue , Transcrição Gênica , Triglicerídeos/sangue
18.
J Biol Chem ; 286(23): 20423-30, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21502324

RESUMO

HMGCS2 (hydroxymethylglutaryl CoA synthase 2), the gene that regulates ketone body production, is barely expressed in cultured cell lines. In this study, we restored HMGCS2 expression and activity in HepG2 cells, thus showing that the wild type enzyme can induce fatty acid ß-oxidation (FAO) and ketogenesis, whereas a catalytically inactive mutant C166A did not generate either process. Peroxisome proliferator-activated receptor (PPAR) α expression also induces fatty acid ß-oxidation and endogenous HMGCS2 expression. Interestingly, PPARα-mediated induction was abolished when HMGCS2 expression was down-regulated by RNAi. These results indicate that HMGCS2 expression is both sufficient and necessary to the control of fatty acid oxidation in these cells. Next, we examined the expression pattern of several PPARα target genes in this now "ketogenic" HepG2 cell line. FGF21 (fibroblast growth factor 21) expression was specifically induced by HMGCS2 activity or by the inclusion of the oxidized form of ketone bodies (acetoacetate) in the culture medium. This effect was blunted by SirT1 (sirtuin 1) RNAi, so we propose a SirT1-dependent mechanism for FGF21 induction by acetoacetate. These data suggest a novel feed-forward mechanism by which HMGCS2 could regulate adaptive metabolic responses during fasting. This mechanism could be physiologically relevant, because fasting-mediated induction of liver FGF21 was dependent on SirT1 activity in vivo.


Assuntos
Ácidos Graxos/metabolismo , Fatores de Crescimento de Fibroblastos/biossíntese , Regulação da Expressão Gênica/fisiologia , Hidroximetilglutaril-CoA Sintase/metabolismo , Substituição de Aminoácidos , Animais , Jejum/fisiologia , Células Hep G2 , Humanos , Hidroximetilglutaril-CoA Sintase/genética , Corpos Cetônicos/genética , Corpos Cetônicos/metabolismo , Camundongos , Camundongos Knockout , Mutação de Sentido Incorreto , Oxirredução , PPAR alfa/genética , PPAR alfa/metabolismo , Sirtuína 1/genética , Sirtuína 1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA