Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Mol Genet Metab ; 137(4): 342-348, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36335793

RESUMO

GM3 synthase (GM3S) deficiency is a rare neurodevelopmental disorder caused by an inability to synthesize gangliosides, for which there is currently no treatment. Gangliosides are brain-enriched, plasma membrane glycosphingolipids with poorly understood biological functions related to cell adhesion, growth, and receptor-mediated signal transduction. Here, we investigated the effects of GM3S deficiency on metabolism and mitochondrial function in a mouse model. By indirect calorimetry, GM3S knockout mice exhibited increased whole-body respiration and an increased reliance upon carbohydrate as an energy source. 18F-FDG PET confirmed higher brain glucose uptake in knockout mice, and GM3S deficient N41 neuronal cells showed higher glucose utilization in vitro. Brain mitochondria from knockout mice respired at a higher rate on Complex I substrates including pyruvate. This appeared to be due to higher expression of pyruvate dehydrogenase (PDH) and lower phosphorylation of PDH, which would favor pyruvate entry into the mitochondrial TCA cycle. Finally, it was observed that blocking glucose metabolism with the glycolysis inhibitor 2-deoxyglucose reduced seizure intensity in GM3S knockout mice following administration of kainate. In conclusion, GM3S deficiency may be associated with a hypermetabolic phenotype that could promote seizure activity.


Assuntos
Glucose , Sialiltransferases , Animais , Camundongos , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Gangliosídeo G(M3)/metabolismo , Glucose/metabolismo , Camundongos Knockout , Ácido Pirúvico , Convulsões/genética , Sialiltransferases/genética , Sialiltransferases/metabolismo
2.
J Lipid Res ; 62: 100069, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33757734

RESUMO

Long-chain fatty acid oxidation is frequently impaired in primary and systemic metabolic diseases affecting the heart; thus, therapeutically increasing reliance on normally minor energetic substrates, such as ketones and medium-chain fatty acids, could benefit cardiac health. However, the molecular fundamentals of this therapy are not fully known. Here, we explored the ability of octanoate, an eight-carbon medium-chain fatty acid known as an unregulated mitochondrial energetic substrate, to ameliorate cardiac hypertrophy in long-chain fatty acid oxidation-deficient hearts because of carnitine palmitoyltransferase 2 deletion (Cpt2M-/-). CPT2 converts acylcarnitines to acyl-CoAs in the mitochondrial matrix for oxidative bioenergetic metabolism. In Cpt2M-/- mice, high octanoate-ketogenic diet failed to alleviate myocardial hypertrophy, dysfunction, and acylcarnitine accumulation suggesting that this alternative substrate is not sufficiently compensatory for energy provision. Aligning this outcome, we identified a major metabolic distinction between muscles and liver, wherein heart and skeletal muscle mitochondria were unable to oxidize free octanoate, but liver was able to oxidize free octanoate. Liver mitochondria, but not heart or muscle, highly expressed medium-chain acyl-CoA synthetases, potentially enabling octanoate activation for oxidation and circumventing acylcarnitine shuttling. Conversely, octanoylcarnitine was oxidized by liver, skeletal muscle, and heart, with rates in heart 4-fold greater than liver and, in muscles, was not dependent upon CPT2. Together, these data suggest that dietary octanoate cannot rescue CPT2-deficient cardiac disease. These data also suggest the existence of tissue-specific mechanisms for octanoate oxidative metabolism, with liver being independent of free carnitine availability, whereas cardiac and skeletal muscles depend on carnitine but not on CPT2.


Assuntos
Carnitina O-Palmitoiltransferase/deficiência , Erros Inatos do Metabolismo
3.
Biochem Biophys Res Commun ; 527(1): 162-166, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32446361

RESUMO

Dicarboxylic fatty acids, taken as a nutritional supplement or produced endogenously via omega oxidation of monocarboxylic fatty acids, may have therapeutic potential for rare inborn errors of metabolism as well as common metabolic diseases such as type 2 diabetes. Breakdown of dicarboxylic acids yields acetyl-CoA and succinyl-CoA as products, the latter of which is anaplerotic for the TCA cycle. However, little is known about the metabolic pathways responsible for degradation of dicarboxylic acids. Here, we demonstrated with whole-cell fatty acid oxidation assays that both mitochondria and peroxisomes contribute to dicarboxylic acid degradation. Several mitochondrial acyl-CoA dehydrogenases were tested for activity against dicarboxylyl-CoAs. Medium-chain acyl-CoA dehydrogenase (MCAD) exhibited activity with both six and 12 carbon dicarboxylyl-CoAs, and the capacity for dehydrogenation of these substrates was significantly reduced in MCAD knockout mouse liver. However, when dicarboxylic acids were fed to normal mice, the expression of MCAD did not change, while expression of peroxisomal fatty acid oxidation enzymes was greatly upregulated. In conclusion, mitochondrial fatty acid oxidation, and in particular MCAD, contributes to dicarboxylic acid degradation, but feeding dicarboxylic acids induces only the peroxisomal pathway.


Assuntos
Acil-CoA Desidrogenases/metabolismo , Ácidos Dicarboxílicos/metabolismo , Ácidos Graxos/metabolismo , Mitocôndrias/enzimologia , Animais , Masculino , Camundongos , Camundongos Knockout
4.
Mol Genet Metab ; 131(1-2): 83-89, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32389575

RESUMO

The fatty acid oxidation enzyme long-chain acyl-CoA dehydrogenase (LCAD) is expressed at high levels in human alveolar type II (ATII) cells in the lung. A common polymorphism causing an amino acid substitution (K333Q) was previously linked to a loss of LCAD antigen in the lung tissue in sudden infant death syndrome. However, the effects of the polymorphism on LCAD function has not been tested. The present work evaluated recombinant LCAD K333Q. Compared to wild-type LCAD protein, LCAD K333Q exhibited significantly reduced enzymatic activity. Molecular modeling suggested that K333 is within interacting distance of the essential FAD cofactor, and the K333Q protein showed a propensity to lose FAD. Exogenous FAD only partially rescued the activity of LCAD K333Q. LCAD K333Q protein was less stable than wild-type when incubated at physiological temperatures, likely explaining the observation of dramatically reduced LCAD antigen in primary ATII cells isolated from individuals homozygous for K333Q. Despite the effect of K333Q on activity, stability, and antigen levels, the frequency of the polymorphism was not increased among infants and children with lung disease.


Assuntos
Acil-CoA Desidrogenase de Cadeia Longa/genética , Estabilidade Enzimática/genética , Pneumopatias/genética , Relação Estrutura-Atividade , Acil-CoA Desidrogenase de Cadeia Longa/ultraestrutura , Animais , Criança , Humanos , Lactente , Pulmão/metabolismo , Pulmão/patologia , Pneumopatias/metabolismo , Pneumopatias/patologia , Modelos Moleculares , Oxirredução , Polimorfismo Genético , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia
5.
J Am Soc Nephrol ; 30(12): 2384-2398, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31575700

RESUMO

BACKGROUND: The primary site of damage during AKI, proximal tubular epithelial cells, are highly metabolically active, relying on fatty acids to meet their energy demands. These cells are rich in mitochondria and peroxisomes, the two organelles that mediate fatty acid oxidation. Emerging evidence shows that both fatty acid pathways are regulated by reversible posttranslational modifications, particularly by lysine acylation. Sirtuin 5 (Sirt5), which localizes to both mitochondria and peroxisomes, reverses post-translational lysine acylation on several enzymes involved in fatty acid oxidation. However, the role of the Sirt5 in regulating kidney energy metabolism has yet to be determined. METHODS: We subjected male Sirt5-deficient mice (either +/- or -/-) and wild-type controls, as well as isolated proximal tubule cells, to two different AKI models (ischemia-induced or cisplatin-induced AKI). We assessed kidney function and injury with standard techniques and measured fatty acid oxidation by the catabolism of 14C-labeled palmitate to 14CO2. RESULTS: Sirt5 was highly expressed in proximal tubular epithelial cells. At baseline, Sirt5 knockout (Sirt5-/- ) mice had modestly decreased mitochondrial function but significantly increased fatty acid oxidation, which was localized to the peroxisome. Although no overt kidney phenotype was observed in Sirt5-/- mice, Sirt5-/- mice had significantly improved kidney function and less tissue damage compared with controls after either ischemia-induced or cisplatin-induced AKI. This coincided with higher peroxisomal fatty acid oxidation compared with mitochondria fatty acid oxidation in the Sirt5-/- proximal tubular epithelial cells. CONCLUSIONS: Our findings indicate that Sirt5 regulates the balance of mitochondrial versus peroxisomal fatty acid oxidation in proximal tubular epithelial cells to protect against injury in AKI. This novel mechanism might be leveraged for developing AKI therapies.


Assuntos
Injúria Renal Aguda/metabolismo , Ácidos Graxos/metabolismo , Túbulos Renais Proximais/metabolismo , Mitocôndrias/metabolismo , Peroxissomos/metabolismo , Sirtuínas/fisiologia , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/patologia , Animais , Cisplatino/toxicidade , Rim/irrigação sanguínea , Masculino , Camundongos , Camundongos Knockout , Oxirredução , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Sirtuínas/deficiência , Sirtuínas/genética
6.
J Biol Chem ; 292(24): 10068-10086, 2017 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-28432125

RESUMO

Hepatocellular carcinoma (HCC) is a common cancer that frequently overexpresses the c-Myc (Myc) oncoprotein. Using a mouse model of Myc-induced HCC, we studied the metabolic, biochemical, and molecular changes accompanying HCC progression, regression, and recurrence. These involved altered rates of pyruvate and fatty acid ß-oxidation and the likely re-directing of glutamine into biosynthetic rather than energy-generating pathways. Initial tumors also showed reduced mitochondrial mass and differential contributions of electron transport chain complexes I and II to respiration. The uncoupling of complex II's electron transport function from its succinate dehydrogenase activity also suggested a mechanism by which Myc generates reactive oxygen species. RNA sequence studies revealed an orderly progression of transcriptional changes involving pathways pertinent to DNA damage repair, cell cycle progression, insulin-like growth factor signaling, innate immunity, and further metabolic re-programming. Only a subset of functions deregulated in initial tumors was similarly deregulated in recurrent tumors thereby indicating that the latter can "normalize" some behaviors to suit their needs. An interactive and freely available software tool was developed to allow continued analyses of these and other transcriptional profiles. Collectively, these studies define the metabolic, biochemical, and molecular events accompanyingHCCevolution, regression, and recurrence in the absence of any potentially confounding therapies.


Assuntos
Carcinoma Hepatocelular/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/metabolismo , Fígado/metabolismo , Neoplasias Experimentais/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Regulação para Cima , Animais , Carcinogênese , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/prevenção & controle , Reparo do DNA , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Complexo II de Transporte de Elétrons/genética , Complexo II de Transporte de Elétrons/metabolismo , Feminino , Perfilação da Expressão Gênica , Inativação Gênica , Humanos , Fígado/patologia , Masculino , Camundongos Transgênicos , Renovação Mitocondrial , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/fisiopatologia , Recidiva Local de Neoplasia/prevenção & controle , Neoplasias Experimentais/patologia , Neoplasias Experimentais/prevenção & controle , Proteínas Proto-Oncogênicas c-myc/genética , Espécies Reativas de Oxigênio/metabolismo , Carga Tumoral
7.
J Biol Chem ; 292(24): 10239-10249, 2017 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-28458255

RESUMO

SIRT5 is a lysine desuccinylase known to regulate mitochondrial fatty acid oxidation and the urea cycle. Here, SIRT5 was observed to bind to cardiolipin via an amphipathic helix on its N terminus. In vitro, succinyl-CoA was used to succinylate liver mitochondrial membrane proteins. SIRT5 largely reversed the succinyl-CoA-driven lysine succinylation. Quantitative mass spectrometry of SIRT5-treated membrane proteins pointed to the electron transport chain, particularly Complex I, as being highly targeted for desuccinylation by SIRT5. Correspondingly, SIRT5-/- HEK293 cells showed defects in both Complex I- and Complex II-driven respiration. In mouse liver, SIRT5 expression was observed to localize strictly to the periportal hepatocytes. However, homogenates prepared from whole SIRT5-/- liver did show reduced Complex II-driven respiration. The enzymatic activities of Complex II and ATP synthase were also significantly reduced. Three-dimensional modeling of Complex II suggested that several SIRT5-targeted lysine residues lie at the protein-lipid interface of succinate dehydrogenase subunit B. We postulate that succinylation at these sites may disrupt Complex II subunit-subunit interactions and electron transfer. Lastly, SIRT5-/- mice, like humans with Complex II deficiency, were found to have mild lactic acidosis. Our findings suggest that SIRT5 is targeted to protein complexes on the inner mitochondrial membrane via affinity for cardiolipin to promote respiratory chain function.


Assuntos
Cardiolipinas/metabolismo , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Hepatócitos/enzimologia , Modelos Moleculares , Processamento de Proteína Pós-Traducional , Sirtuínas/metabolismo , Substituição de Aminoácidos , Animais , Cardiolipinas/química , Complexo I de Transporte de Elétrons/metabolismo , Complexo II de Transporte de Elétrons/metabolismo , Células HEK293 , Hepatócitos/metabolismo , Humanos , Lisina/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias Hepáticas/enzimologia , Mitocôndrias Hepáticas/metabolismo , Membranas Mitocondriais/enzimologia , Membranas Mitocondriais/metabolismo , Mutação , Transporte Proteico , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Sirtuínas/química , Sirtuínas/genética
8.
Biochem Biophys Res Commun ; 497(2): 700-704, 2018 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-29458021

RESUMO

We previously showed that the mitochondrial fatty acid oxidation enzyme long-chain acyl-CoA dehydrogenase (LCAD) is expressed in alveolar type II pneumocytes and that LCAD-/- mice have altered breathing mechanics and surfactant defects. Here, we hypothesized that LCAD-/- mice would be susceptible to influenza infection. Indeed, LCAD-/- mice demonstrated increased mortality following infection with 2009 pandemic influenza (A/CA/07/09). However, the mortality was not due to increased lung injury, as inflammatory cell counts, viral titers, and histology scores all showed non-significant trends toward milder injury in LCAD-/- mice. To confirm this, LCAD-/- were infected with a second, mouse-adapted H1N1 virus (A/PR/8/34), to which they responded with significantly less lung injury. While both strains become increasingly hypoglycemic over the first week post-infection, LCAD-/- mice lose body weight more rapidly than wild-type mice. Surprisingly, while acutely fasted LCAD-/- mice develop hepatic steatosis, influenza-infected LCAD-/- mice do not. They do, however, become more hypothermic than wild-type mice and demonstrate increased blood lactate values. We conclude that LCAD-/- mice succumb to influenza from bioenergetic starvation, likely due to increased reliance upon glucose for energy.


Assuntos
Acil-CoA Desidrogenase de Cadeia Longa/genética , Técnicas de Silenciamento de Genes , Vírus da Influenza A Subtipo H1N1/fisiologia , Pulmão/patologia , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/patologia , Animais , Peso Corporal , Feminino , Hipotermia/etiologia , Hipotermia/genética , Hipotermia/patologia , Hipotermia/virologia , Pulmão/virologia , Camundongos , Camundongos Knockout , Infecções por Orthomyxoviridae/complicações , Infecções por Orthomyxoviridae/virologia
9.
Biochem Biophys Res Commun ; 482(2): 346-351, 2017 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-27856258

RESUMO

The metabolic effects of salicylates are poorly understood. This study investigated the effects of aspirin on fatty acid oxidation. Aspirin increased mitochondrial long-chain fatty acid oxidation, but inhibited peroxisomal fatty acid oxidation, in two different cell lines. Aspirin increased mitochondrial protein acetylation and was found to be a stronger acetylating agent in vitro than acetyl-CoA. However, aspirin-induced acetylation did not alter the activity of fatty acid oxidation proteins, and knocking out the mitochondrial deacetylase SIRT3 did not affect the induction of long-chain fatty acid oxidation by aspirin. Aspirin did not change oxidation of medium-chain fatty acids, which can freely traverse the mitochondrial membrane. Together, these data indicate that aspirin does not directly alter mitochondrial matrix fatty acid oxidation enzymes, but most likely exerts its effects at the level of long-chain fatty acid transport into mitochondria. The drive on mitochondrial fatty acid oxidation may be a compensatory response to altered mitochondrial morphology and inhibited electron transport chain function, both of which were observed after 24 h incubation of cells with aspirin. These studies provide insight into the pathophysiology of Reye Syndrome, which is known to be triggered by aspirin ingestion in patients with fatty acid oxidation disorders.


Assuntos
Aspirina/administração & dosagem , Ácidos Graxos/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Anti-Inflamatórios não Esteroides/administração & dosagem , Respiração Celular/fisiologia , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Taxa de Depuração Metabólica/efeitos dos fármacos , Oxirredução
10.
J Biol Chem ; 289(15): 10668-10679, 2014 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-24591516

RESUMO

Long-chain acyl-CoA dehydrogenase (LCAD) is a mitochondrial fatty acid oxidation enzyme whose expression in humans is low or absent in organs known to utilize fatty acids for energy such as heart, muscle, and liver. This study demonstrates localization of LCAD to human alveolar type II pneumocytes, which synthesize and secrete pulmonary surfactant. The physiological role of LCAD and the fatty acid oxidation pathway in lung was subsequently studied using LCAD knock-out mice. Lung fatty acid oxidation was reduced in LCAD(-/-) mice. LCAD(-/-) mice demonstrated reduced pulmonary compliance, but histological examination of lung tissue revealed no obvious signs of inflammation or pathology. The changes in lung mechanics were found to be due to pulmonary surfactant dysfunction. Large aggregate surfactant isolated from LCAD(-/-) mouse lavage fluid had significantly reduced phospholipid content as well as alterations in the acyl chain composition of phosphatidylcholine and phosphatidylglycerol. LCAD(-/-) surfactant demonstrated functional abnormalities when subjected to dynamic compression-expansion cycling on a constrained drop surfactometer. Serum albumin, which has been shown to degrade and inactivate pulmonary surfactant, was significantly increased in LCAD(-/-) lavage fluid, suggesting increased epithelial permeability. Finally, we identified two cases of sudden unexplained infant death where no lung LCAD antigen was detectable. Both infants were homozygous for an amino acid changing polymorphism (K333Q). These findings for the first time identify the fatty acid oxidation pathway and LCAD in particular as factors contributing to the pathophysiology of pulmonary disease.


Assuntos
Acil-CoA Desidrogenase de Cadeia Longa/deficiência , Erros Inatos do Metabolismo Lipídico/metabolismo , Pneumopatias/etiologia , Surfactantes Pulmonares/metabolismo , Acil-CoA Desidrogenase de Cadeia Longa/metabolismo , Adulto , Animais , Brônquios/metabolismo , Linhagem Celular Tumoral , Coenzima A/metabolismo , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Ácidos Graxos/metabolismo , Feminino , Homozigoto , Humanos , Lactente , Recém-Nascido , Pulmão/metabolismo , Pneumopatias/metabolismo , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oxigênio/metabolismo , Fosfatidilcolinas/química , Fosfatidilgliceróis/química , Polimorfismo Genético , Alvéolos Pulmonares/metabolismo
11.
J Biol Chem ; 288(47): 33837-33847, 2013 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-24121500

RESUMO

Long-chain acyl-CoA dehydrogenase (LCAD) is a key mitochondrial fatty acid oxidation enzyme. We previously demonstrated increased LCAD lysine acetylation in SIRT3 knockout mice concomitant with reduced LCAD activity and reduced fatty acid oxidation. To study the effects of acetylation on LCAD and determine sirtuin 3 (SIRT3) target sites, we chemically acetylated recombinant LCAD. Acetylation impeded substrate binding and reduced catalytic efficiency. Deacetylation with recombinant SIRT3 partially restored activity. Residues Lys-318 and Lys-322 were identified as SIRT3-targeted lysines. Arginine substitutions at Lys-318 and Lys-322 prevented the acetylation-induced activity loss. Lys-318 and Lys-322 flank residues Arg-317 and Phe-320, which are conserved among all acyl-CoA dehydrogenases and coordinate the enzyme-bound FAD cofactor in the active site. We propose that acetylation at Lys-318/Lys-322 causes a conformational change which reduces hydride transfer from substrate to FAD. Medium-chain acyl-CoA dehydrogenase and acyl-CoA dehydrogenase 9, two related enzymes with lysines at positions equivalent to Lys-318/Lys-322, were also efficiently deacetylated by SIRT3 following chemical acetylation. These results suggest that acetylation/deacetylation at Lys-318/Lys-322 is a mode of regulating fatty acid oxidation. The same mechanism may regulate other acyl-CoA dehydrogenases.


Assuntos
Ácidos Graxos/metabolismo , Flavina-Adenina Dinucleotídeo/metabolismo , Mitocôndrias Hepáticas/enzimologia , Sirtuína 3/metabolismo , Acetilação , Acil-CoA Desidrogenase de Cadeia Longa , Animais , Domínio Catalítico/fisiologia , Ácidos Graxos/química , Ácidos Graxos/genética , Flavina-Adenina Dinucleotídeo/química , Flavina-Adenina Dinucleotídeo/genética , Humanos , Camundongos , Camundongos Knockout , Mitocôndrias Hepáticas/genética , Oxirredução , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sirtuína 3/química , Sirtuína 3/genética
12.
J Clin Invest ; 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38687608

RESUMO

Dicarboxylic fatty acids are generated in the liver and kidney in a minor pathway called fatty acid ω-oxidation. The effects of consuming dicarboxylic fatty acids as an alternative source of dietary fat have not been explored. Here, we fed dodecanedioic acid, a 12-carbon dicarboxylic (DC12), to mice at 20% of daily caloric intake for nine weeks. DC12 increased metabolic rate, reduced body fat, reduced liver fat, and improved glucose tolerance. We observed DC12-specific breakdown products in liver, kidney, muscle, heart, and brain, indicating that oral DC12 escaped first-pass liver metabolism and was utilized by many tissues. In tissues expressing the "a" isoform of acyl-CoA oxidase-1 (ACOX1), a key peroxisomal fatty acid oxidation enzyme, DC12 was chain shortened to the TCA cycle intermediate succinyl-CoA. In tissues with low peroxisomal fatty acid oxidation capacity, DC12 was oxidized by mitochondria. In vitro, DC12 was catabolized even by adipose tissue and was not stored intracellularly. We conclude that DC12 and other dicarboxylic acids may be useful for combatting obesity and for treating metabolic disorders.

13.
Sci Rep ; 10(1): 18367, 2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-33110171

RESUMO

Medium-chain triglycerides (MCT), containing C8-C12 fatty acids, are used to treat several pediatric disorders and are widely consumed as a nutritional supplement. Here, we investigated the role of the sirtuin deacylase Sirt5 in MCT metabolism by feeding Sirt5 knockout mice (Sirt5KO) high-fat diets containing either C8/C10 fatty acids or coconut oil, which is rich in C12, for five weeks. Coconut oil, but not C8/C10 feeding, induced periportal macrovesicular steatosis in Sirt5KO mice. 14C-C12 degradation was significantly reduced in Sirt5KO liver. This decrease was localized to the mitochondrial ß-oxidation pathway, as Sirt5KO mice exhibited no change in peroxisomal C12 ß-oxidation. Endoplasmic reticulum ω-oxidation, a minor fatty acid degradation pathway known to be stimulated by C12 accumulation, was increased in Sirt5KO liver. Mice lacking another mitochondrial C12 oxidation enzyme, long-chain acyl-CoA dehydrogenase (LCAD), also developed periportal macrovesicular steatosis when fed coconut oil, confirming that defective mitochondrial C12 oxidation is sufficient to induce the steatosis phenotype. Sirt5KO liver exhibited normal LCAD activity but reduced mitochondrial acyl-CoA synthetase activity with C12. These studies reveal a role for Sirt5 in regulating the hepatic response to MCT and may shed light into the pathogenesis of periportal steatosis, a hallmark of human pediatric non-alcoholic fatty liver disease.


Assuntos
Ácidos Graxos/metabolismo , Mitocôndrias Hepáticas/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Sirtuínas/genética , Acil-CoA Desidrogenase de Cadeia Longa/metabolismo , Animais , Óleo de Coco/administração & dosagem , Gorduras na Dieta/administração & dosagem , Feminino , Masculino , Camundongos , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/genética , Oxirredução , Triglicerídeos/metabolismo
14.
Redox Biol ; 26: 101253, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31234015

RESUMO

Fatty acid oxidation (FAO)-driven H2O2 has been shown to be a major source of oxidative stress in several tissues and disease states. Here, we established that the mitochondrial flavoprotein long-chain acyl-CoA dehydrogenase (LCAD), which catalyzes a key step in mitochondrial FAO, directly produces H2O2in vitro by leaking electrons to oxygen. Kinetic analysis of recombinant human LCAD showed that it produces H2O2 15-fold faster than the related mitochondrial enzyme very long-chain acyl-CoA dehydrogenase (VLCAD), but 50-fold slower than a bona fide peroxisomal acyl-CoA oxidase. The rate of H2O2 formation by human LCAD is slow compared to its activity as a dehydrogenase (about 1%). However, expression of hLCAD in HepG2 cells is sufficient to significantly increase H2O2 in the presence of fatty acids. Liver mitochondria from LCAD-/- mice, but not VLCAD-/- mice, produce significantly less H2O2 during incubation with fatty acids. Finally, we observe highest LCAD expression in human liver, followed by kidney, lung, and pancreas. Based on our data, we propose that the presence of LCAD drives H2O2 formation in response to fatty acids in these tissues.


Assuntos
Acil-CoA Desidrogenase de Cadeia Longa/metabolismo , Acil-CoA Oxidase/metabolismo , Ácidos Graxos/metabolismo , Peróxido de Hidrogênio/metabolismo , Fígado/enzimologia , Mitocôndrias Hepáticas/enzimologia , Acil-CoA Desidrogenase de Cadeia Longa/genética , Acil-CoA Oxidase/genética , Animais , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Células Hep G2 , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Rim/enzimologia , Cinética , Pulmão/enzimologia , Masculino , Camundongos , Camundongos Knockout , Especificidade de Órgãos , Oxirredução , Estresse Oxidativo , Pâncreas/enzimologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
16.
Cancer Res ; 77(21): 5795-5807, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28883002

RESUMO

Rapidly proliferating cells increase glycolysis at the expense of oxidative phosphorylation (oxphos) to generate sufficient levels of glycolytic intermediates for use as anabolic substrates. The pyruvate dehydrogenase complex (PDC) is a critical mitochondrial enzyme that catalyzes pyruvate's conversion to acetyl coenzyme A (AcCoA), thereby connecting these two pathways in response to complex energetic, enzymatic, and metabolic cues. Here we utilized a mouse model of hepatocyte-specific PDC inactivation to determine the need for this metabolic link during normal hepatocyte regeneration and malignant transformation. In PDC "knockout" (KO) animals, the long-term regenerative potential of hepatocytes was unimpaired, and growth of aggressive experimental hepatoblastomas was only modestly slowed in the face of 80%-90% reductions in AcCoA and significant alterations in the levels of key tricarboxylic acid (TCA) cycle intermediates and amino acids. Overall, oxphos activity in KO livers and hepatoblastoma was comparable with that of control counterparts, with evidence that metabolic substrate abnormalities were compensated for by increased mitochondrial mass. These findings demonstrate that the biochemical link between glycolysis and the TCA cycle can be completely severed without affecting normal or neoplastic proliferation, even under the most demanding circumstances. Cancer Res; 77(21); 5795-807. ©2017 AACR.


Assuntos
Proliferação de Células , Ciclo do Ácido Cítrico , Glicólise , Hepatócitos/metabolismo , Proteínas Mitocondriais/metabolismo , Acetilcoenzima A/metabolismo , Animais , Células Cultivadas , Feminino , Hepatoblastoma/genética , Hepatoblastoma/metabolismo , Hepatoblastoma/patologia , Hepatócitos/citologia , Immunoblotting , Camundongos Knockout , Proteínas Mitocondriais/genética , Fosforilação Oxidativa , Complexo Piruvato Desidrogenase/genética , Complexo Piruvato Desidrogenase/metabolismo , Ácido Pirúvico/metabolismo , Análise de Sobrevida , Espectrometria de Massas em Tandem
17.
PLoS One ; 10(3): e0122297, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25811481

RESUMO

SIRT3 and SIRT5 have been shown to regulate mitochondrial fatty acid oxidation but the molecular mechanisms behind the regulation are lacking. Here, we demonstrate that SIRT3 and SIRT5 both target human very long-chain acyl-CoA dehydrogenase (VLCAD), a key fatty acid oxidation enzyme. SIRT3 deacetylates and SIRT5 desuccinylates K299 which serves to stabilize the essential FAD cofactor in the active site. Further, we show that VLCAD binds strongly to cardiolipin and isolated mitochondrial membranes via a domain near the C-terminus containing lysines K482, K492, and K507. Acetylation or succinylation of these residues eliminates binding of VLCAD to cardiolipin. SIRT3 deacetylates K507 while SIRT5 desuccinylates K482, K492, and K507. Sirtuin deacylation of recombinant VLCAD rescues membrane binding. Endogenous VLCAD from SIRT3 and SIRT5 knockout mouse liver shows reduced binding to cardiolipin. Thus, SIRT3 and SIRT5 promote fatty acid oxidation by converging upon VLCAD to promote its activity and membrane localization. Regulation of cardiolipin binding by reversible lysine acylation is a novel mechanism that is predicted to extrapolate to other metabolic proteins that localize to the inner mitochondrial membrane.


Assuntos
Acil-CoA Desidrogenase de Cadeia Longa/metabolismo , Cardiolipinas/metabolismo , Sirtuína 3/metabolismo , Sirtuínas/metabolismo , Acil-CoA Desidrogenase de Cadeia Longa/química , Acilação , Sequência de Aminoácidos , Animais , Sítios de Ligação , Cardiolipinas/química , Domínio Catalítico , Ativação Enzimática , Flavina-Adenina Dinucleotídeo/química , Flavina-Adenina Dinucleotídeo/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias Hepáticas/metabolismo , Membranas Mitocondriais/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Alinhamento de Sequência , Sirtuína 3/química , Sirtuína 3/genética , Sirtuínas/química , Sirtuínas/genética
18.
J Matern Fetal Neonatal Med ; 26(2): 150-4, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22928498

RESUMO

OBJECTIVE: To measure the effect labor exerts on fatty acid (FA) oxidation in term human placentas, and to compare enzymes expression and activity between placenta and liver. METHODS: Placental samples were collected: (a) scheduled non-labored cesarean section and (b) normal vaginal delivery at or beyond 37 weeks. Long and medium-chain FA oxidation were measured using (3)H-labeled FA, ATP concentration was measured via commercial kit. Activity and expression levels of 11 FA enzymes were measured and results compared to both human and mouse liver. RESULTS: Placentas undergoing labor had significantly decreased palmitate oxidation and ATP levels. Octanoic acid oxidation was 10-fold higher than palmitic acid oxidation. No difference in expression or activity level was detected between the groups. CONCLUSION: Term human placentas express all the enzymes required to oxidize FA, at a rate 20-fold lower than liver. FA Oxidation is not likely an important placental energy source during labor. Further work is needed to determine the functionality of this pathway in placenta.


Assuntos
Ácidos Graxos/metabolismo , Trabalho de Parto/metabolismo , Placenta/metabolismo , Acil-CoA Desidrogenase/metabolismo , Animais , Feminino , Humanos , Metabolismo dos Lipídeos , Fígado/metabolismo , Camundongos , Oxirredução , Gravidez , Trítio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA