Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Am Acad Dermatol ; 78(3 Suppl 1): S53-S62, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29248518

RESUMO

The Janus kinase-signal transducer and activator of transcription pathway is a conserved master regulator of immunity and myeloproliferation. Advanced understanding of this pathway has led to development of targeted inhibitors of Janus kinases (Jakinibs). As a class, JAK inhibitors effectively treat a multitude of hematologic and inflammatory diseases. Given such success, use of JAK inhibitors for mitigation of atopic dermatitis is under active investigation. Herein, we review the evolving data on the safety and efficacy of JAK inhibitors in treatment of atopic dermatitis. Although it is still early in the study of JAK inhibitors for atopic dermatitis, evidence identifies JAK inhibitors as effective alternatives to conventional therapies. Nonetheless, multiple large safety and efficacy trials are needed before widespread use of JAK inhibitors can be advocated for atopic dermatitis.


Assuntos
Dermatite Atópica/tratamento farmacológico , Inibidores de Janus Quinases/uso terapêutico , Terapia de Alvo Molecular/métodos , Canais de Cátion TRPV/genética , Adulto , Animais , Ensaios Clínicos Fase II como Assunto , Ensaios Clínicos Fase III como Assunto , Dermatite Atópica/diagnóstico , Dermatite Atópica/genética , Feminino , Humanos , Inibidores de Janus Quinases/farmacologia , Masculino , Segurança do Paciente , Prognóstico , Resultado do Tratamento
2.
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
3.
Am J Physiol Endocrinol Metab ; 307(2): E176-85, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24865983

RESUMO

Peroxisome proliferator activated receptor-α (PPARα) is a master transcriptional regulator of hepatic metabolism and mediates the adaptive response to fasting. Here, we demonstrate the roles for PPARα in hepatic metabolic adaptations to birth. Like fasting, nutrient supply is abruptly altered at birth when a transplacental source of carbohydrates is replaced by a high-fat, low-carbohydrate milk diet. PPARα-knockout (KO) neonatal mice exhibit relative hypoglycemia due to impaired conversion of glycerol to glucose. Although hepatic expression of fatty acyl-CoA dehydrogenases is imparied in PPARα neonates, these animals exhibit normal blood acylcarnitine profiles. Furthermore, quantitative metabolic fate mapping of the medium-chain fatty acid [(13)C]octanoate in neonatal mouse livers revealed normal contribution of this fatty acid to the hepatic TCA cycle. Interestingly, octanoate-derived carbon labeled glucose uniquely in livers of PPARα-KO neonates. Relative hypoketonemia in newborn PPARα-KO animals could be mechanistically linked to a 50% decrease in de novo hepatic ketogenesis from labeled octanoate. Decreased ketogenesis was associated with diminished mRNA and protein abundance of the fate-committing ketogenic enzyme mitochondrial 3-hydroxymethylglutaryl-CoA synthase (HMGCS2) and decreased protein abundance of the ketogenic enzyme ß-hydroxybutyrate dehydrogenase 1 (BDH1). Finally, hepatic triglyceride and free fatty acid concentrations were increased 6.9- and 2.7-fold, respectively, in suckling PPARα-KO neonates. Together, these findings indicate a primary defect of gluconeogenesis from glycerol and an important role for PPARα-dependent ketogenesis in the disposal of hepatic fatty acids during the neonatal period.


Assuntos
Gluconeogênese/genética , Corpos Cetônicos/metabolismo , Fígado/metabolismo , PPAR alfa/genética , Animais , Animais Recém-Nascidos , Ácidos Graxos/metabolismo , Glicerol/metabolismo , Hipoglicemia/genética , Hipoglicemia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução
4.
Am J Physiol Endocrinol Metab ; 304(4): E363-74, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23233542

RESUMO

During states of low carbohydrate intake, mammalian ketone body metabolism transfers energy substrates originally derived from fatty acyl chains within the liver to extrahepatic organs. We previously demonstrated that the mitochondrial enzyme coenzyme A (CoA) transferase [succinyl-CoA:3-oxoacid CoA transferase (SCOT), encoded by nuclear Oxct1] is required for oxidation of ketone bodies and that germline SCOT-knockout (KO) mice die within 48 h of birth because of hyperketonemic hypoglycemia. Here, we use novel transgenic and tissue-specific SCOT-KO mice to demonstrate that ketone bodies do not serve an obligate energetic role within highly ketolytic tissues during the ketogenic neonatal period or during starvation in the adult. Although transgene-mediated restoration of myocardial CoA transferase in germline SCOT-KO mice is insufficient to prevent lethal hyperketonemic hypoglycemia in the neonatal period, mice lacking CoA transferase selectively within neurons, cardiomyocytes, or skeletal myocytes are all viable as neonates. Like germline SCOT-KO neonatal mice, neonatal mice with neuronal CoA transferase deficiency exhibit increased cerebral glycolysis and glucose oxidation, and, while these neonatal mice exhibit modest hyperketonemia, they do not develop hypoglycemia. As adults, tissue-specific SCOT-KO mice tolerate starvation, exhibiting only modestly increased hyperketonemia. Finally, metabolic analysis of adult germline Oxct1(+/-) mice demonstrates that global diminution of ketone body oxidation yields hyperketonemia, but hypoglycemia emerges only during a protracted state of low carbohydrate intake. Together, these data suggest that, at the tissue level, ketone bodies are not a required energy substrate in the newborn period or during starvation, but rather that integrated ketone body metabolism mediates adaptation to ketogenic nutrient states.


Assuntos
Envelhecimento , Coenzima A-Transferases/metabolismo , Corpos Cetônicos/metabolismo , Cetose/fisiopatologia , Músculo Esquelético/enzimologia , Miócitos Cardíacos/enzimologia , Neurônios/enzimologia , Adaptação Fisiológica , Animais , Animais Recém-Nascidos , Restrição Calórica/efeitos adversos , Coenzima A-Transferases/biossíntese , Coenzima A-Transferases/genética , Heterozigoto , Hipoglicemia/etiologia , Corpos Cetônicos/sangue , Cetose/sangue , Cetose/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Especificidade de Órgãos , Oxirredução
5.
Am J Physiol Heart Circ Physiol ; 304(8): H1060-76, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23396451

RESUMO

Ketone bodies are metabolized through evolutionarily conserved pathways that support bioenergetic homeostasis, particularly in brain, heart, and skeletal muscle when carbohydrates are in short supply. The metabolism of ketone bodies interfaces with the tricarboxylic acid cycle, ß-oxidation of fatty acids, de novo lipogenesis, sterol biosynthesis, glucose metabolism, the mitochondrial electron transport chain, hormonal signaling, intracellular signal transduction pathways, and the microbiome. Here we review the mechanisms through which ketone bodies are metabolized and how their signals are transmitted. We focus on the roles this metabolic pathway may play in cardiovascular disease states, the bioenergetic benefits of myocardial ketone body oxidation, and prospective interactions among ketone body metabolism, obesity, metabolic syndrome, and atherosclerosis. Ketone body metabolism is noninvasively quantifiable in humans and is responsive to nutritional interventions. Therefore, further investigation of this pathway in disease models and in humans may ultimately yield tailored diagnostic strategies and therapies for specific pathological states.


Assuntos
Doenças Cardiovasculares/metabolismo , Corpos Cetônicos/metabolismo , Animais , Cardiomiopatias/metabolismo , Coenzima A-Transferases/metabolismo , Ácidos Graxos/metabolismo , Humanos , Hidroximetilglutaril-CoA Sintase/metabolismo , Lipogênese/fisiologia , Fígado/metabolismo , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Hepáticas/metabolismo , Miocárdio/metabolismo , Obesidade/metabolismo , Oxirredução , Transdução de Sinais
6.
Cureus ; 15(6): e40582, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37469822

RESUMO

Disseminated superficial actinic porokeratosis (DSAP) is a disorder of abnormal keratinization for which there is no standard treatment. Treatment modalities that have traditionally been utilized with varying success include ablative therapies, topical pharmacologic treatments, surgical excision, and retinoids. The underlying pathophysiology of DSAP is secondary to genetic mutations in the mevalonate biosynthesis pathway, and thus topical lovastatin/cholesterol presents a promising treatment modality for this condition. We present a case of familial DSAP successfully treated with topical lovastatin/cholesterol gel and provide a brief review of the existing literature surrounding this novel therapy.

7.
J Biol Chem ; 286(9): 6902-10, 2011 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-21209089

RESUMO

To compensate for the energetic deficit elicited by reduced carbohydrate intake, mammals convert energy stored in ketone bodies to high energy phosphates. Ketone bodies provide fuel particularly to brain, heart, and skeletal muscle in states that include starvation, adherence to low carbohydrate diets, and the neonatal period. Here, we use novel Oxct1(-/-) mice, which lack the ketolytic enzyme succinyl-CoA:3-oxo-acid CoA-transferase (SCOT), to demonstrate that ketone body oxidation is required for postnatal survival in mice. Although Oxct1(-/-) mice exhibit normal prenatal development, all develop ketoacidosis, hypoglycemia, and reduced plasma lactate concentrations within the first 48 h of birth. In vivo oxidation of (13)C-labeled ß-hydroxybutyrate in neonatal Oxct1(-/-) mice, measured using NMR, reveals intact oxidation to acetoacetate but no contribution of ketone bodies to the tricarboxylic acid cycle. Accumulation of acetoacetate yields a markedly reduced ß-hydroxybutyrate:acetoacetate ratio of 1:3, compared with 3:1 in Oxct1(+) littermates. Frequent exogenous glucose administration to actively suckling Oxct1(-/-) mice delayed, but could not prevent, lethality. Brains of newborn SCOT-deficient mice demonstrate evidence of adaptive energy acquisition, with increased phosphorylation of AMP-activated protein kinase α, increased autophagy, and 2.4-fold increased in vivo oxidative metabolism of [(13)C]glucose. Furthermore, [(13)C]lactate oxidation is increased 1.7-fold in skeletal muscle of Oxct1(-/-) mice but not in brain. These results indicate the critical metabolic roles of ketone bodies in neonatal metabolism and suggest that distinct tissues exhibit specific metabolic responses to loss of ketone body oxidation.


Assuntos
Coenzima A-Transferases/genética , Coenzima A-Transferases/metabolismo , Metabolismo Energético/fisiologia , Homeostase/fisiologia , Corpos Cetônicos/metabolismo , Adaptação Fisiológica/fisiologia , Animais , Animais Recém-Nascidos , Autofagia/fisiologia , Glicemia/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Membrana Celular/metabolismo , Hipoglicemia/metabolismo , Hipoglicemia/patologia , Cetose/metabolismo , Cetose/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Ressonância Magnética Nuclear Biomolecular , Oxirredução
8.
J Bacteriol ; 193(21): 5936-49, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21873491

RESUMO

Yersinia pestis CO92 has 12 open reading frames encoding putative conventional autotransporters (yaps), nine of which appear to produce functional proteins. Here, we demonstrate the ability of the Yap proteins to localize to the cell surface of both Escherichia coli and Yersinia pestis and show that a subset of these proteins undergoes processing by bacterial surface omptins to be released into the supernatant. Numerous autotransporters have been implicated in pathogenesis, suggesting a role for the Yaps as virulence factors in Y. pestis. Using the C57BL/6 mouse models of bubonic and pneumonic plague, we determined that all of these genes are transcribed in the lymph nodes during bubonic infection and in the lungs during pneumonic infection, suggesting a role for the Yaps during mammalian infection. In vitro transcription studies did not identify a particular environmental stimulus responsible for transcriptional induction. The primary sequences of the Yaps reveal little similarity to any characterized autotransporters; however, two of the genes are present in operons, suggesting that the proteins encoded in these operons may function together. Further work aims to elucidate the specific functions of the Yaps and clarify the contributions of these proteins to Y. pestis pathogenesis.


Assuntos
Regulação Bacteriana da Expressão Gênica , Proteínas de Membrana Transportadoras/biossíntese , Peste/microbiologia , Yersinia pestis/genética , Yersinia pestis/metabolismo , Animais , Modelos Animais de Doenças , Escherichia coli/genética , Escherichia coli/metabolismo , Perfilação da Expressão Gênica , Pulmão/microbiologia , Linfonodos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Doenças dos Roedores/microbiologia , Serina Endopeptidases/metabolismo , Fatores de Virulência/biossíntese
9.
J Biol Chem ; 285(32): 24447-56, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20529848

RESUMO

Heart muscle is metabolically versatile, converting energy stored in fatty acids, glucose, lactate, amino acids, and ketone bodies. Here, we use mouse models in ketotic nutritional states (24 h of fasting and a very low carbohydrate ketogenic diet) to demonstrate that heart muscle engages a metabolic response that limits ketone body utilization. Pathway reconstruction from microarray data sets, gene expression analysis, protein immunoblotting, and immunohistochemical analysis of myocardial tissue from nutritionally modified mouse models reveal that ketotic states promote transcriptional suppression of the key ketolytic enzyme, succinyl-CoA:3-oxoacid CoA transferase (SCOT; encoded by Oxct1), as well as peroxisome proliferator-activated receptor alpha-dependent induction of the key ketogenic enzyme HMGCS2. Consistent with reduction of SCOT, NMR profiling demonstrates that maintenance on a ketogenic diet causes a 25% reduction of myocardial (13)C enrichment of glutamate when (13)C-labeled ketone bodies are delivered in vivo or ex vivo, indicating reduced procession of ketones through oxidative metabolism. Accordingly, unmetabolized substrate concentrations are higher within the hearts of ketogenic diet-fed mice challenged with ketones compared with those of chow-fed controls. Furthermore, reduced ketone body oxidation correlates with failure of ketone bodies to inhibit fatty acid oxidation. These results indicate that ketotic nutrient environments engage mechanisms that curtail ketolytic capacity, controlling the utilization of ketone bodies in ketotic states.


Assuntos
Miocárdio/metabolismo , Animais , Isótopos de Carbono/química , Coenzima A-Transferases/metabolismo , Imuno-Histoquímica/métodos , Corpos Cetônicos/química , Cetonas/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Miócitos Cardíacos/citologia , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Ratos
10.
Am J Physiol Gastrointest Liver Physiol ; 300(6): G956-67, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21454445

RESUMO

Low-carbohydrate diets are used to manage obesity, seizure disorders, and malignancies of the central nervous system. These diets create a distinctive, but incompletely defined, cellular, molecular, and integrated metabolic state. Here, we determine the systemic and hepatic effects of long-term administration of a very low-carbohydrate, low-protein, and high-fat ketogenic diet, serially comparing these effects to a high-simple-carbohydrate, high-fat Western diet and a low-fat, polysaccharide-rich control chow diet in C57BL/6J mice. Longitudinal measurement of body composition, serum metabolites, and intrahepatic fat content, using in vivo magnetic resonance spectroscopy, reveals that mice fed the ketogenic diet over 12 wk remain lean, euglycemic, and hypoinsulinemic but accumulate hepatic lipid in a temporal pattern very distinct from animals fed the Western diet. Ketogenic diet-fed mice ultimately develop systemic glucose intolerance, hepatic endoplasmic reticulum stress, steatosis, cellular injury, and macrophage accumulation, but surprisingly insulin-induced hepatic Akt phosphorylation and whole-body insulin responsiveness are not impaired. Moreover, whereas hepatic Pparg mRNA abundance is augmented by both high-fat diets, each diet confers splice variant specificity. The distinctive nutrient milieu created by long-term administration of this low-carbohydrate, low-protein ketogenic diet in mice evokes unique signatures of nonalcoholic fatty liver disease and whole-body glucose homeostasis.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Dieta com Restrição de Carboidratos/efeitos adversos , Dieta Cetogênica/efeitos adversos , Retículo Endoplasmático/metabolismo , Fígado Gorduroso/etiologia , Inflamação/etiologia , Fígado/metabolismo , Estresse Fisiológico , Análise de Variância , Animais , Biomarcadores/sangue , Glicemia/metabolismo , Composição Corporal , Dieta com Restrição de Proteínas , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/metabolismo , Retículo Endoplasmático/patologia , Ingestão de Energia , Ácidos Graxos não Esterificados/sangue , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Fígado Gorduroso/fisiopatologia , Regulação da Expressão Gênica , Intolerância à Glucose/etiologia , Intolerância à Glucose/metabolismo , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Inflamação/fisiopatologia , Mediadores da Inflamação/metabolismo , Insulina/sangue , Resistência à Insulina , Fígado/patologia , Fígado/fisiopatologia , Espectroscopia de Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , PPAR gama/genética , PPAR gama/metabolismo , Fatores de Tempo , Triglicerídeos/sangue , Resposta a Proteínas não Dobradas
11.
Cureus ; 12(10): e10879, 2020 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-33178531

RESUMO

Cutaneous squamous cell carcinoma (cSCC) typically arises from a malignant proliferation of keratinocytes. It is the second most common cancer in the United States and typically affects older white men. Risk factors for cSCC include ultraviolet radiation exposure, light skin tone, and immunosuppression. Although metastasis in cSCC is rare, primary tumor characteristics such as location, size, and depth of invasion, among others, can help risk-stratify lesions for local recurrence, metastatic events, and death. We present a case of primary cutaneous metastatic squamous cell carcinoma masquerading as a cyst on the left temple of a 73-year-old Caucasian man following numerous treatments of cryotherapy to an ipsilateral helical lesion.

12.
Cureus ; 12(4): e7562, 2020 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-32382464

RESUMO

Cutaneous sclerosis occurs in association with a variety of systemic diseases, including hematologic malignancy, plasma cell dyscrasias, solid organ tumors, and other systemic autoimmune conditions. Herein, we present a unique case of morphea/lichen sclerosus overlap arising in association with aplastic anemia. To expand upon this rare case, we also review the literature surrounding paraneoplastic sclerosing skin disorders. A 53-year-old man presented with a 13-month history of progressive and generalized skin changes. Exam revealed irregular, hypopigmented indurated plaques with focal areas of scale on the bilateral axillae and hips, as well as hyperpigmented brown papules and plaques on the back. Laboratory evaluation revealed pancytopenia and positive anti-nuclear antibody (1:160). Bone marrow biopsy demonstrated hypocellular marrow consistent with aplastic anemia. Furthermore, skin biopsies revealed lichen sclerosus overlying superficial morphea, consistent with a paraneoplastic sclerodermoid-like eruption. While preparations for hematologic-directed therapies were made, skin-directed therapy with a combination topical steroids and topical calcineurin inhibitors was initiated. Eosinophilic fasciitis and scleroderma have been linked to aplastic anemia, and herein, we expand upon this phenomenon by presenting our case of generalized plaque morphea/lichen sclerosus overlap arising in the setting of aplastic anemia. Dermatologists must be aware of this rare association in order to identify precocious hematologic disease.

13.
Cureus ; 11(2): e4021, 2019 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-31007979

RESUMO

Leukemia cutis, or infiltration of leukemic cells into the skin, occurs rarely in B-cell acute lymphocytic leukemia (ALL). Herein, we have described a rare, precocious presentation of B-cell ALL presenting as indurated facial plaques in a 69-year-old man. Biopsy of the facial plaques revealed precursor B-cell leukemia/lymphoma in the skin and prompted urgent hematologic-oncologic evaluation. Bone marrow biopsy yielded a final diagnosis of B-cell ALL. The patient underwent induction therapy, and at the last available follow-up, a matched unrelated donor transplant was planned.

16.
J Clin Invest ; 124(12): 5175-90, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25347470

RESUMO

Nonalcoholic fatty liver disease (NAFLD) spectrum disorders affect approximately 1 billion individuals worldwide. However, the drivers of progressive steatohepatitis remain incompletely defined. Ketogenesis can dispose of much of the fat that enters the liver, and dysfunction in this pathway could promote the development of NAFLD. Here, we evaluated mice lacking mitochondrial 3-hydroxymethylglutaryl CoA synthase (HMGCS2) to determine the role of ketogenesis in preventing diet-induced steatohepatitis. Antisense oligonucleotide-induced loss of HMGCS2 in chow-fed adult mice caused mild hyperglycemia, increased hepatic gluconeogenesis from pyruvate, and augmented production of hundreds of hepatic metabolites, a suite of which indicated activation of the de novo lipogenesis pathway. High-fat diet feeding of mice with insufficient ketogenesis resulted in extensive hepatocyte injury and inflammation, decreased glycemia, deranged hepatic TCA cycle intermediate concentrations, and impaired hepatic gluconeogenesis due to sequestration of free coenzyme A (CoASH). Supplementation of the CoASH precursors pantothenic acid and cysteine normalized TCA intermediates and gluconeogenesis in the livers of ketogenesis-insufficient animals. Together, these findings indicate that ketogenesis is a critical regulator of hepatic acyl-CoA metabolism, glucose metabolism, and TCA cycle function in the absorptive state and suggest that ketogenesis may modulate fatty liver disease.


Assuntos
Gorduras na Dieta/efeitos adversos , Glucose/metabolismo , Hiperglicemia/induzido quimicamente , Hiperglicemia/metabolismo , Hepatopatia Gordurosa não Alcoólica/induzido quimicamente , Hepatopatia Gordurosa não Alcoólica/metabolismo , Acil Coenzima A/genética , Acil Coenzima A/metabolismo , Animais , Ciclo do Ácido Cítrico/efeitos dos fármacos , Ciclo do Ácido Cítrico/genética , Gluconeogênese/efeitos dos fármacos , Gluconeogênese/genética , Glucose/genética , Hidroximetilglutaril-CoA Sintase/deficiência , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Hiperglicemia/genética , Hiperglicemia/patologia , Hipoglicemia/genética , Hipoglicemia/metabolismo , Hipoglicemia/patologia , Masculino , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/metabolismo , Erros Inatos do Metabolismo/patologia , Camundongos , Camundongos Mutantes , Doenças Mitocondriais/genética , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/patologia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa