Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 783
Filtrar
1.
Sci Rep ; 14(1): 17942, 2024 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-39095513

RESUMO

Glycogen storage, conversion and utilization in astrocytes play an important role in brain energy metabolism. The conversion of glycogen to lactate through glycolysis occurs through the coordinated activities of various enzymes and inhibition of this process can impair different brain processes including formation of long-lasting memories. To replenish depleted glycogen stores, astrocytes undergo glycogen synthesis, a cellular process that has been shown to require transcription and translation during specific stimulation paradigms. However, the detail nuclear signaling mechanisms and transcriptional regulation during glycogen synthesis in astrocytes remains to be explored. In this report, we study the molecular mechanisms of vasoactive intestinal peptide (VIP)-induced glycogen synthesis in astrocytes. VIP is a potent neuropeptide that triggers glycogenolysis followed by glycogen synthesis in astrocytes. We show evidence that VIP-induced glycogen synthesis requires CREB-mediated transcription that is calcium dependent and requires conventional Protein Kinase C but not Protein Kinase A. In parallel to CREB activation, we demonstrate that VIP also triggers nuclear accumulation of the CREB coactivator CRTC2 in astrocytic nuclei. Transcriptome profiles of VIP-induced astrocytes identified robust CREB transcription, including a subset of genes linked to glucose and glycogen metabolism. Finally, we demonstrate that VIP-induced glycogen synthesis shares similar as well as distinct molecular signatures with glucose-induced glycogen synthesis, including the requirement of CREB-mediated transcription. Overall, our data demonstrates the importance of CREB-mediated transcription in astrocytes during stimulus-driven glycogenesis.


Assuntos
Astrócitos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Glicogênio , Peptídeo Intestinal Vasoativo , Astrócitos/metabolismo , Glicogênio/metabolismo , Glicogênio/biossíntese , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Peptídeo Intestinal Vasoativo/metabolismo , Transcrição Gênica , Células Cultivadas , Proteína Quinase C/metabolismo , Regulação da Expressão Gênica , Camundongos , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Núcleo Celular/metabolismo
2.
J Clin Endocrinol Metab ; 107(3): e1193-e1203, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-34665856

RESUMO

CONTEXT: Exercise blunts the effect of beta2-agonists on peripheral glucose uptake and energy expenditure. Whether such attenuation extends into recovery is unknown. OBJECTIVE: To examine the effect of a beta2-agonist on leg glucose uptake and metabolic rate in recovery from exercise. METHODS: Using leg arteriovenous balance technique and analyses of thigh muscle biopsies, we investigated the effect of a beta2-agonist (24 mg of oral salbutamol) vs placebo on leg glucose, lactate, and oxygen exchange before and during quadriceps exercise, and 0.5 to 5 hours in recovery from quadriceps exercise, as well as on muscle glycogen resynthesis and activity in recovery. Twelve healthy, lean, young men participated. RESULTS: Before exercise, leg glucose uptake was 0.42 ±â€…0.12 and 0.20 ±â€…0.02 mmol × min-1 (mean ±â€…SD) for salbutamol and placebo (P = .06), respectively, while leg oxygen consumption was around 2-fold higher (P < .01) for salbutamol than for placebo (25 ±â€…3 vs 14 ±â€…1 mL × min-1). No treatment differences were observed in leg glucose uptake, lactate release, and oxygen consumption during exercise. But in recovery, cumulated leg glucose uptake, lactate release, and oxygen consumption was 21 mmol (95% CI 18-24, P = .018), 19 mmol (95% CI 16-23, P < .01), and 1.8 L (95% CI 1.6-2.0, P < .01) higher for salbutamol than for placebo, respectively. Muscle glycogen content was around 30% lower (P < .01) for salbutamol than for placebo in recovery, whereas no treatment differences were observed in muscle glycogen resynthesis or glycogen synthase activity. CONCLUSION: Exercise blunts the effect of beta2-agonist salbutamol on leg glucose uptake, but this attenuation diminishes in recovery. Salbutamol increases leg lactate release in recovery, which may relate to glycolytic trafficking due to excessive myocellular glucose uptake.


Assuntos
Albuterol/administração & dosagem , Exercício Físico , Glucose/metabolismo , Glicogênio/biossíntese , Músculo Esquelético/efeitos dos fármacos , Adulto , Biópsia , Metabolismo Energético , Glucose/análise , Glicogênio/análise , Glicólise/efeitos dos fármacos , Voluntários Saudáveis , Humanos , Ácido Láctico/análise , Ácido Láctico/metabolismo , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Coxa da Perna
3.
Plant Cell Physiol ; 63(1): 135-147, 2022 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-34698867

RESUMO

In photoautotrophic Synechocystis sp. PCC 6803, NADPH is generated from photosynthesis and utilized in various metabolism, including the biosynthesis of glyceraldehyde 3-phosphate (the upstream substrate for carbon metabolism), poly(3-hydroxybutyrate) (PHB), photosynthetic pigments, and hydrogen gas (H2). Redirecting NADPH flow from one biosynthesis pathway to another has yet to be studied. Synechocystis's H2 synthesis, one of the pathways consuming NAD(P)H, was disrupted by the inactivation of hoxY and hoxH genes encoding the two catalytic subunits of hydrogenase. Such inactivation with a complete disruption of H2 synthesis led to 1.4-, 1.9-, and 2.1-fold increased cellular NAD(P)H levels when cells were cultured in normal medium (BG11), the medium without nitrate (-N), and the medium without phosphate (-P), respectively. After 49-52 d of cultivation in BG11 (when the nitrogen source in the media was depleted), the cells with disrupted H2 synthesis had 1.3-fold increased glycogen level compared to wild type of 83-85% (w/w dry weight), the highest level reported for cyanobacterial glycogen. The increased glycogen content observed by transmission electron microscopy was correlated with the increased levels of glucose 6-phosphate and glucose 1-phosphate, the two substrates in glycogen synthesis. Disrupted H2 synthesis also enhanced PHB accumulation up to 1.4-fold under -P and 1.6-fold under -N and increased levels of photosynthetic pigments (chlorophyll a, phycocyanin, and allophycocyanin) by 1.3- to 1.5-fold under BG11. Thus, disrupted H2 synthesis increased levels of NAD(P)H, which may be utilized for the biosynthesis of glycogen, PHB, and pigments. This strategy might be applicable for enhancing other biosynthetic pathways that utilize NAD(P)H.


Assuntos
Clorofila/biossíntese , Glicogênio/biossíntese , Hidrogênio/metabolismo , Hidroxibutiratos/metabolismo , NADP/metabolismo , Synechocystis/química , Synechocystis/genética , Synechocystis/metabolismo , Clorofila/genética , Regulação da Expressão Gênica de Plantas , Genes de Plantas , Glicogênio/genética , Redes e Vias Metabólicas , NADP/genética
4.
JCI Insight ; 6(19)2021 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-34622807

RESUMO

The signaling mechanisms by which dietary fat and cholesterol signals regulate central pathways of glucose homeostasis are not completely understood. By using a hepatocyte-specific PKCß-deficient (PKCßHep-/-) mouse model, we demonstrated the role of hepatic PKCß in slowing disposal of glucose overload by suppressing glycogenesis and increasing hepatic glucose output. PKCßHep-/- mice exhibited lower plasma glucose under the fed condition, modestly improved systemic glucose tolerance and mildly suppressed gluconeogenesis, increased hepatic glycogen accumulation and synthesis due to elevated glucokinase expression and activated glycogen synthase (GS), and suppressed glucose-6-phosphatase expression compared with controls. These events were independent of hepatic AKT/GSK-3α/ß signaling and were accompanied by increased HNF-4α transactivation, reduced FoxO1 protein abundance, and elevated expression of GS targeting protein phosphatase 1 regulatory subunit 3C in the PKCßHep-/- liver compared with controls. The above data strongly imply that hepatic PKCß deficiency causes hypoglycemia postprandially by promoting glucose phosphorylation via upregulating glucokinase and subsequently redirecting more glucose-6-phosphate to glycogen via activating GS. In summary, hepatic PKCß has a unique and essential ability to induce a coordinated response that negatively affects glycogenesis at multiple levels under physiological postprandial conditions, thereby integrating nutritional fat intake with dysregulation of glucose homeostasis.


Assuntos
Glicemia/metabolismo , Gorduras na Dieta , Glicogênio/biossíntese , Fígado/metabolismo , Proteína Quinase C beta/genética , Animais , Colesterol na Dieta , Proteína Forkhead Box O1/metabolismo , Glucoquinase/metabolismo , Gluconeogênese/genética , Glucose-6-Fosfatase/metabolismo , Glicogênio/metabolismo , Glicogênio Sintase/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Camundongos , Camundongos Knockout , Período Pós-Prandial/genética , Proteína Quinase C beta/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais
5.
J Ethnopharmacol ; 281: 114556, 2021 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-34438036

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Aloe vera (L.) Burm.f. is an ancient medicinal plant that belongs to the family Asphodelaceae. It has a rich source of bioactive constituents such as carbohydrates, polyphenols, peptides, sterols and tannins, etc. Aloe vera has multiple biological activities such as anti-inflammatory, antioxidant and antidiabetic activity etc. AIM OF THE STUDY: The present study investigated the antidiabetic mechanism of Aloe vera carbohydrate fraction (AVCF) and aimed to provide insights into the regulation of carbohydrate metabolism enzymes in glucose homeostasis. MATERIALS AND METHODS: The antidiabetic effect of AVCF was evaluated using α-amylase, α-glucosidase inhibition, glucose diffusion and glucose uptake assay. The in vitro AVCF effect on insulin secretion, cell proliferation and inflammatory markers were determined using streptozotocin-induced oxidative stress on RIN-m5F cells. Streptozotocin-induced male Wistar diabetic rats were treated for 21 days with AVCF (54 mg/kg bw). The in vivo AVCF effect was measured on fasting plasma glucose, insulin, glucagon, hexokinase, glycogen synthase and glucose-6-phosphatase, levels in diabetic rats. Histopathological studies for organ-specific effects in the pancreas, liver and small intestine were also conducted. RESULTS: AVCF-treated RIN-m5F cells significantly increased BrdU levels, with insulin secretion, and decreased TNF-α, IL-6 and nitric oxide levels. AVCF treated streptozotocin-induced diabetic rats showed significantly decreased fasting plasma glucose, glucagon and glucose-6-phosphatase levels with a concomitant increase in insulin, hexokinase, and glycogen synthase levels and, glycogen content. These findings corroborate with the improved hepatic glycogen content in the PAS stained histological section of the liver of AVCF treated diabetic rats. CONCLUSION: These results suggest that CF of Aloe vera improved glucose metabolism by activation of glycogenesis and down-regulation of gluconeogenesis thereby, maintaining glucose homeostasis. Hence, AVCF can be used as an alternative medicine in the alleviation of diabetes mellitus symptoms.


Assuntos
Aloe/química , Carboidratos/farmacologia , Glucose/metabolismo , Glicogênio/biossíntese , Fígado/efeitos dos fármacos , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citocinas/sangue , Citocinas/genética , Citocinas/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Regulação para Baixo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Gluconeogênese/efeitos dos fármacos , Inflamação/sangue , Inflamação/metabolismo , Insulina/sangue , Ilhotas Pancreáticas/efeitos dos fármacos , Fígado/metabolismo , Masculino , Óxido Nítrico/sangue , Distribuição Aleatória , Ratos , Ratos Wistar
6.
Proc Natl Acad Sci U S A ; 118(31)2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34330832

RESUMO

UDP-glucose pyrophosphorylase 2 (UGP2), the enzyme that synthesizes uridine diphosphate (UDP)-glucose, rests at the convergence of multiple metabolic pathways, however, the role of UGP2 in tumor maintenance and cancer metabolism remains unclear. Here, we identify an important role for UGP2 in the maintenance of pancreatic ductal adenocarcinoma (PDAC) growth in both in vitro and in vivo tumor models. We found that transcription of UGP2 is directly regulated by the Yes-associated protein 1 (YAP)-TEA domain transcription factor (TEAD) complex, identifying UGP2 as a bona fide YAP target gene. Loss of UGP2 leads to decreased intracellular glycogen levels and defects in N-glycosylation targets that are important for the survival of PDACs, including the epidermal growth factor receptor (EGFR). These critical roles of UGP2 in cancer maintenance, metabolism, and protein glycosylation may offer insights into therapeutic options for otherwise intractable PDACs.


Assuntos
Carcinoma Ductal Pancreático/enzimologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Glicogênio/biossíntese , Neoplasias Pancreáticas/enzimologia , UTP-Glucose-1-Fosfato Uridililtransferase/metabolismo , Animais , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Glicosilação , Humanos , Camundongos , Camundongos Nus , Neoplasias Experimentais , Neoplasias Pancreáticas/patologia , Fatores de Transcrição de Domínio TEA/genética , Fatores de Transcrição de Domínio TEA/metabolismo , UTP-Glucose-1-Fosfato Uridililtransferase/genética , Proteínas de Sinalização YAP/genética , Proteínas de Sinalização YAP/metabolismo
7.
J Cell Mol Med ; 25(5): 2714-2724, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33523562

RESUMO

Abnormal glucose metabolism may contribute to cancer progression. As a member of the CRK (v-crk sarcoma virus CT10 oncogene homologue) adapter protein family, CRKL (CRK-like) associated with the development and progression of various tumours. However, the exact role and underlying mechanism of CRKL on energy metabolism remain unknown. In this study, we investigated the effect of CRKL on glucose metabolism of hepatocarcinoma cells. CRKL and PI3K were found to be overexpressed in both hepatocarcinoma cells and tissues; meanwhile, CRKL up-regulation was positively correlated with PI3K up-regulation. Functional investigations revealed that CRKL overexpression promoted glucose uptake, lactate production and glycogen synthesis of hepatocarcinoma cells by up-regulating glucose transporters 1 (GLUT1), hexokinase II (HKII) expression and down-regulating glycogen synthase kinase 3ß (GSK3ß) expression. Mechanistically, CRKL promoted glucose metabolism of hepatocarcinoma cells via enhancing the CRKL-PI3K/Akt-GLUT1/HKII-glucose uptake, CRKL-PI3K/Akt-HKII-glucose-lactate production and CRKL-PI3K/Akt-Gsk3ß-glycogen synthesis. We demonstrate CRKL facilitates HCC malignancy via enhancing glucose uptake, lactate production and glycogen synthesis through PI3K/Akt pathway. It provides interesting fundamental clues to CRKL-related carcinogenesis through glucose metabolism and offers novel therapeutic strategies for hepatocarcinoma.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Carcinoma Hepatocelular/etiologia , Carcinoma Hepatocelular/metabolismo , Glucose/metabolismo , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Suscetibilidade a Doenças , Regulação Neoplásica da Expressão Gênica , Glicogênio/biossíntese , Humanos , Neoplasias Hepáticas/patologia , Proteômica/métodos , Transdução de Sinais
8.
Phytomedicine ; 83: 153473, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33513558

RESUMO

BACKGROUND: Diabetes mellitus is a chronic metabolic disease characterized by increased blood glucose levels. In order to lower blood glucose, it is important to stimulate glucose uptake and glycogen synthesis in the muscle. (E)-5-hydroxy-7-methoxy-3-(2'-hydroxybenzyl)-4-chromanone (HM-chromanone), a constituent isolated from Portulaca oleracea L., exhibits anti-diabetic effects; however, its mechanisms are not yet clearly understood on glucose uptake and glycogen synthesis in muscle cells. PURPOSE: In the present study, we examined the effects of HM-chromanone on glucose uptake into L6 skeletal muscle cells and elucidated the underlying mechanisms. METHODS: The effects of HM-chromanone on glucose uptake into L6 skeletal muscle cells were assessed by 2-Deoxyglucose uptake assay. Western blot analysis was carried out to elucidate the underlying molecular mechanisms. RESULTS: We found that HM-chromanone promoted glucose uptake into L6 skeletal muscle cells in a dose-dependent manner. Moreover, HM-chromanone induced the phosphorylation of IRS-1Tyr612 and AKTSer473, and the activation of PI3K. HM-chromanone also stimulated the phosphorylation of AMPKThr172, AS160Thr642, TBC1D1Ser237, and ACC via the CaMKKß pathway. Furthermore, HM-chromanone increased glycogen synthesis through the inactivation of glycogen synthase kinase 3 α/ß. CONCLUSION: The results of this study indicate that HM-chromanone stimulates glucose uptake through the activation of the PI3K/AKT and CaMKKß-AMPK pathways and glycogen synthesis via the GSK3 α/ß pathway in L6 skeletal muscle cells.


Assuntos
Flavonoides/farmacologia , Glucose/metabolismo , Glicogênio/biossíntese , Músculo Esquelético/efeitos dos fármacos , Portulaca/química , Animais , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Células Cultivadas , Desoxiglucose/metabolismo , Glicogênio/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos
9.
Nature ; 590(7844): 122-128, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33473210

RESUMO

Ageing is characterized by the development of persistent pro-inflammatory responses that contribute to atherosclerosis, metabolic syndrome, cancer and frailty1-3. The ageing brain is also vulnerable to inflammation, as demonstrated by the high prevalence of age-associated cognitive decline and Alzheimer's disease4-6. Systemically, circulating pro-inflammatory factors can promote cognitive decline7,8, and in the brain, microglia lose the ability to clear misfolded proteins that are associated with neurodegeneration9,10. However, the underlying mechanisms that initiate and sustain maladaptive inflammation with ageing are not well defined. Here we show that in ageing mice myeloid cell bioenergetics are suppressed in response to increased signalling by the lipid messenger prostaglandin E2 (PGE2), a major modulator of inflammation11. In ageing macrophages and microglia, PGE2 signalling through its EP2 receptor promotes the sequestration of glucose into glycogen, reducing glucose flux and mitochondrial respiration. This energy-deficient state, which drives maladaptive pro-inflammatory responses, is further augmented by a dependence of aged myeloid cells on glucose as a principal fuel source. In aged mice, inhibition of myeloid EP2 signalling rejuvenates cellular bioenergetics, systemic and brain inflammatory states, hippocampal synaptic plasticity and spatial memory. Moreover, blockade of peripheral myeloid EP2 signalling is sufficient to restore cognition in aged mice. Our study suggests that cognitive ageing is not a static or irrevocable condition but can be reversed by reprogramming myeloid glucose metabolism to restore youthful immune functions.


Assuntos
Envelhecimento/metabolismo , Disfunção Cognitiva/prevenção & controle , Células Mieloides/metabolismo , Adulto , Idoso , Envelhecimento/efeitos dos fármacos , Envelhecimento/genética , Animais , Respiração Celular , Células Cultivadas , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/genética , Dinoprostona/metabolismo , Metabolismo Energético , Glucose/metabolismo , Glicogênio/biossíntese , Glicogênio/metabolismo , Humanos , Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Transtornos da Memória/tratamento farmacológico , Camundongos , Microglia/efeitos dos fármacos , Microglia/imunologia , Microglia/metabolismo , Mitocôndrias/metabolismo , Células Mieloides/imunologia , Receptores de Prostaglandina E Subtipo EP2/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP2/deficiência , Receptores de Prostaglandina E Subtipo EP2/genética , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Memória Espacial/efeitos dos fármacos
10.
Am J Physiol Cell Physiol ; 319(6): C1151-C1157, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33026831

RESUMO

Branched-chain amino acids (BCAAs) are essential for critical metabolic processes; however, recent studies have associated elevated plasma BCAA levels with increased risk of insulin resistance. Using skeletal muscle cells, we aimed to determine whether continued exposure of high extracellular BCAA would result in impaired insulin signaling and whether the compound sodium phenylbutyrate (PB), which induces BCAA metabolism, would lower extracellular BCAA, thereby alleviating their potentially inhibitory effects on insulin-mediated signaling. Prolonged exposure of elevated BCAA to cells resulted in impaired insulin receptor substrate 1/AKT signaling and insulin-stimulated glycogen synthesis. PB significantly reduced media BCAA and branched-chain keto acid concentrations and increased phosphorylation of AKT [+2.0 ± 0.1-fold; P < 0.001 versus without (-)PB] and AS160 (+3.2 ± 0.2-fold; P < 0.001 versus -PB); however, insulin-stimulated glycogen synthesis was further reduced upon PB treatment. Continued exposure of high BCAA resulted in impaired intracellular insulin signaling and glycogen synthesis, and while forcing BCAA catabolism using PB resulted in increases in proteins important for regulating glucose uptake, PB did not prevent the impairments in glycogen synthesis with BCAA exposure.


Assuntos
Aminoácidos de Cadeia Ramificada/metabolismo , Glicogênio/biossíntese , Resistência à Insulina/fisiologia , Insulina/metabolismo , Músculo Esquelético/metabolismo , Animais , Linhagem Celular , Proteínas Substratos do Receptor de Insulina/metabolismo , Camundongos , Células Musculares/metabolismo , Fenilbutiratos/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
11.
J Pharmacol Exp Ther ; 373(1): 81-91, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32024803

RESUMO

Oleoylethanolamide (OEA) is an endogenous peroxisome proliferator-activated receptor α (PPARα) agonist that acts on the peripheral control of energy metabolism. However, its therapeutic potential and related mechanisms in hepatic glucose metabolism under type 2 diabetes mellitus (T2DM) are not clear. Here, OEA treatment markedly improved glucose homeostasis in a PPARα-independent manner. OEA efficiently promoted glycogen synthesis and suppressed gluconeogenesis in mouse primary hepatocytes and liver tissue. OEA enhanced hepatic glycogen synthesis and inhibited gluconeogenesis via liver kinase B1 (LKB1)/5' AMP-activated protein kinase (AMPK) signaling pathways. PPARα was not involved in the roles of OEA in the LKB1/AMPK pathways. We found that OEA exerts its antidiabetic effect by increasing glycogenesis and decreasing gluconeogenesis via the LKB1/AMPK pathway. The ability of OEA to control hepatic LKB1/AMPK pathways may serve as a novel therapeutic approach for the treatment of T2DM. SIGNIFICANCE STATEMENT: Oleoylethanolamide (OEA) exerted a potent antihyperglycemic effect in a peroxisome proliferator-activated receptor α-independent manner. OEA played an antihyperglycemic role primarily via regulation of hepatic glycogen synthesis and gluconeogenesis. The main molecular mechanism of OEA in regulating liver glycometabolism is activating the liver kinase B1/5' AMP-activated protein kinase signaling pathways.


Assuntos
Diabetes Mellitus Tipo 2/sangue , Endocanabinoides/farmacologia , Gluconeogênese/fisiologia , Glicogênio/biossíntese , Fígado/metabolismo , Ácidos Oleicos/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 2/induzido quimicamente , Diabetes Mellitus Tipo 2/tratamento farmacológico , Endocanabinoides/uso terapêutico , Gluconeogênese/efeitos dos fármacos , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Ácidos Oleicos/uso terapêutico , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
12.
In Vitro Cell Dev Biol Anim ; 56(1): 75-84, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31845076

RESUMO

Porcine hepatocytes are a promising option for xenotransplantation in light of the critical shortage of orthotopic donor livers. Because primary hepatocytes have limited ability to proliferate in vitro, several immortalized hepatocyte lines have been established. However, these cells have typically been generated using a virus-dependent transfection methodology and express viral oncogenes that introduce potential risks in clinical applications. In our study, we established immortalized porcine neonatal hepatocytes by introduction of a plasmid-based hTERT gene expression system by electroporation, without the use of viral components. We detected stable expression of hTERT by RT-PCR and Western blot. The immortalized hepatocytes exhibit a high growth rate, but retain the normal morphology of freshly isolated primary hepatocytes. To date, these immortalized hepatocytes have been expanded for over 80 passages. In addition, no significant differences were detected in glycogen synthesis, secretion of serum albumin, or lipid accumulation between the primary hepatocytes and our immortalized hepatocytes. The cells also exhibit serum-dependent growth and have no capacity for anchorage-independent growth in vitro, demonstrating that they have not been transformed in vitro. Our immortalized porcine hepatocytes will be useful for elucidating the pathogenesis of liver disease and developing efficient treatments. Furthermore, these immortalized hepatocytes may provide a safer source of cells for application in xenotransplantation, compared with immortalized cells generated using viral components.


Assuntos
Técnicas de Cultura de Células/métodos , Hepatócitos/citologia , Vírus/metabolismo , Animais , Animais Recém-Nascidos , Linhagem Celular Transformada , Proliferação de Células , Forma Celular , Glicogênio/biossíntese , Hepatócitos/metabolismo , Humanos , Lipídeos/química , Masculino , Albumina Sérica/metabolismo , Suínos , Telomerase/metabolismo
13.
Cell Metab ; 30(5): 903-916.e7, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31523006

RESUMO

Nuclear glycogen was first documented in the early 1940s, but its role in cellular physiology remained elusive. In this study, we utilized pure nuclei preparations and stable isotope tracers to define the origin and metabolic fate of nuclear glycogen. Herein, we describe a key function for nuclear glycogen in epigenetic regulation through compartmentalized pyruvate production and histone acetylation. This pathway is altered in human non-small cell lung cancers, as surgical specimens accumulate glycogen in the nucleus. We demonstrate that the decreased abundance of malin, an E3 ubiquitin ligase, impaired nuclear glycogenolysis by preventing the nuclear translocation of glycogen phosphorylase and causing nuclear glycogen accumulation. Re-introduction of malin in lung cancer cells restored nuclear glycogenolysis, increased histone acetylation, and decreased growth of cancer cells transplanted into mice. This study uncovers a previously unknown role for glycogen metabolism in the nucleus and elucidates another mechanism by which cellular metabolites control epigenetic regulation.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Núcleo Celular/metabolismo , Glicogenólise/genética , Histonas/metabolismo , Neoplasias Pulmonares/metabolismo , Células A549 , Acetilação , Animais , Carbono/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Glicogênio/biossíntese , Glicogênio Fosforilase/metabolismo , Células HEK293 , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Knockout , Camundongos Nus , Transfecção , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
14.
Biochem J ; 476(14): 2059-2092, 2019 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-31366571

RESUMO

The evolution of metabolic pathways is a major force behind natural selection. In the spotlight of such process lies the structural evolution of the enzymatic machinery responsible for the central energy metabolism. Specifically, glycogen metabolism has emerged to allow organisms to save available environmental surplus of carbon and energy, using dedicated glucose polymers as a storage compartment that can be mobilized at future demand. The origins of such adaptive advantage rely on the acquisition of an enzymatic system for the biosynthesis and degradation of glycogen, along with mechanisms to balance the assembly and disassembly rate of this polysaccharide, in order to store and recover glucose according to cell energy needs. The first step in the classical bacterial glycogen biosynthetic pathway is carried out by the adenosine 5'-diphosphate (ADP)-glucose pyrophosphorylase. This allosteric enzyme synthesizes ADP-glucose and acts as a point of regulation. The second step is carried out by the glycogen synthase, an enzyme that generates linear α-(1→4)-linked glucose chains, whereas the third step catalyzed by the branching enzyme produces α-(1→6)-linked glucan branches in the polymer. Two enzymes facilitate glycogen degradation: glycogen phosphorylase, which functions as an α-(1→4)-depolymerizing enzyme, and the debranching enzyme that catalyzes the removal of α-(1→6)-linked ramifications. In this work, we rationalize the structural basis of glycogen metabolism in bacteria to the light of the current knowledge. We describe and discuss the remarkable progress made in the understanding of the molecular mechanisms of substrate recognition and product release, allosteric regulation and catalysis of all those enzymes.


Assuntos
Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Glucose-1-Fosfato Adenililtransferase/metabolismo , Glicogênio Sintase/metabolismo , Glicogênio/biossíntese , Regulação Alostérica
15.
Hum Gene Ther ; 30(10): 1263-1273, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31319709

RESUMO

Glycogen storage diseases (GSDs) type I (GSDI) and type III (GSDIII), the most frequent hepatic GSDs, are due to defects in glycogen metabolism, mainly in the liver. In addition to hypoglycemia and liver pathology, renal, myeloid, or muscle complications affect GSDI and GSDIII patients. Currently, patient management is based on dietary treatment preventing severe hypoglycemia and increasing the lifespan of patients. However, most of the patients develop long-term pathologies. In the past years, gene therapy for GSDI has generated proof of concept for hepatic GSDs. This resulted in a recent clinical trial of adeno-associated virus (AAV)-based gene replacement for GSDIa. However, the current limitations of AAV-mediated gene transfer still represent a challenge for successful gene therapy in GSDI and GSDIII. Indeed, transgene loss over time was observed in GSDI liver, possibly due to the degeneration of hepatocytes underlying the physiopathology of both GSDI and GSDIII and leading to hepatic tumor development. Moreover, multitissue targeting requires high vector doses to target nonpermissive tissues such as muscle and kidney. Interestingly, recent pharmacological interventions or dietary regimen aiming at the amelioration of the hepatocyte abnormalities before the administration of gene therapy demonstrated improved efficacy in GSDs. In this review, we describe the advances in gene therapy and the limitations to be overcome to achieve efficient and safe gene transfer in GSDs.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Glucose-6-Fosfatase/genética , Doença de Depósito de Glicogênio Tipo III/terapia , Doença de Depósito de Glicogênio Tipo I/terapia , Hipoglicemia/terapia , Animais , Ensaios Clínicos como Assunto , Dependovirus/metabolismo , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Glucose-6-Fosfatase/metabolismo , Glicogênio/biossíntese , Doença de Depósito de Glicogênio Tipo I/enzimologia , Doença de Depósito de Glicogênio Tipo I/genética , Doença de Depósito de Glicogênio Tipo I/patologia , Doença de Depósito de Glicogênio Tipo III/enzimologia , Doença de Depósito de Glicogênio Tipo III/genética , Doença de Depósito de Glicogênio Tipo III/patologia , Hepatócitos/enzimologia , Hepatócitos/patologia , Humanos , Hipoglicemia/enzimologia , Hipoglicemia/genética , Hipoglicemia/patologia , Fígado/enzimologia , Fígado/patologia , Transgenes
16.
Proc Natl Acad Sci U S A ; 116(8): 2977-2986, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30728291

RESUMO

Tuberous sclerosis complex (TSC) is an autosomal dominant syndrome that causes tumor formation in multiple organs. TSC is caused by inactivating mutations in the genes encoding TSC1/2, negative regulators of the mammalian target of rapamycin complex 1 (mTORC1). Diminished TSC function is associated with excess glycogen storage, but the causative mechanism is unknown. By studying human and mouse cells with defective or absent TSC2, we show that complete loss of TSC2 causes an increase in glycogen synthesis through mTORC1 hyperactivation and subsequent inactivation of glycogen synthase kinase 3ß (GSK3ß), a negative regulator of glycogen synthesis. Specific TSC2 pathogenic mutations, however, result in elevated glycogen levels with no changes in mTORC1 or GSK3ß activities. We identify mTORC1-independent lysosomal depletion and impairment of autophagy as the driving causes underlying abnormal glycogen storage in TSC irrespective of the underlying mutation. The defective autophagic degradation of glycogen is associated with abnormal ubiquitination and degradation of essential proteins of the autophagy-lysosome pathway, such as LC3 and lysosomal associated membrane protein 1 and 2 (LAMP1/2) and is restored by the combined use of mTORC1 and Akt pharmacological inhibitors. In complementation to current models that place mTORC1 as the central therapeutic target for TSC pathogenesis, our findings identify mTORC1-independent pathways that are dysregulated in TSC and that should therefore be taken into account in the development of a therapeutic treatment.


Assuntos
Glicogênio Sintase Quinase 3 beta/genética , Glicogênio/biossíntese , Proteína 2 do Complexo Esclerose Tuberosa/genética , Esclerose Tuberosa/genética , Animais , Autofagia/genética , Glicogênio/genética , Humanos , Proteína 1 de Membrana Associada ao Lisossomo/genética , Proteína 2 de Membrana Associada ao Lisossomo/genética , Lisossomos/genética , Lisossomos/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Camundongos , Mutação , Proteólise , Transdução de Sinais , Esclerose Tuberosa/patologia , Ubiquitinação/genética
17.
Dig Dis Sci ; 64(6): 1548-1559, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30627917

RESUMO

BACKGROUND/AIM: The miR-181 family plays an important role in the regulation of various cellular functions. However, whether miR-181b-5p mediates hepatic insulin resistance remains unknown. In this study, we investigated the effect of miR-181b-5p on the regulation of hepatic glycogen synthesis. METHODS: The miR-181b-5p levels in the livers of diabetic mice were detected by real-time PCR. The glycogen levels and AKT/GSK pathway activation were examined in human hepatic L02 cells and HepG2 cells transfected with miR-181b-5p mimic or inhibitor. The potential target genes of miR-181b-5p were evaluated using a luciferase reporter assay and Western blot analysis. EGR1-specific siRNA and pCMV-EGR1 were used to further determine the role of miR-181b-5p in hepatic glycogen synthesis in vitro. Hepatic inhibition of miR-181b-5p in mice was performed using adeno-associated virus 8 (AAV8) vectors by tail intravenous injection. RESULTS: The miR-181b-5p levels were significantly decreased in the serum and livers of diabetic mice as well as the serum of type 2 diabetes patients. Importantly, inhibition of miR-181b-5p expression impaired the AKT/GSK pathway and reduced glycogenesis in hepatocytes. Moreover, upregulation of miR-181b-5p reversed high-glucose-induced suppression of glycogenesis. Further analysis revealed that early growth response 1 (EGR1) was a downstream target of miR-181b-5p. Silencing of EGR1 expression rescued miR-181b-5p inhibition-reduced AKT/GSK pathway activation and glycogenesis in hepatocytes. Hepatic inhibition of miR-181b-5p led to insulin resistance in C57BL/6 J mice. CONCLUSION: We demonstrated that miR-181b-5p contributes to glycogen synthesis by targeting EGR1, thereby regulating PTEN expression to mediate hepatic insulin resistance.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Glicogênio/biossíntese , Resistência à Insulina , Fígado/metabolismo , MicroRNAs/metabolismo , Adulto , Animais , Diabetes Mellitus Tipo 2/genética , Modelos Animais de Doenças , Proteína 1 de Resposta de Crescimento Precoce/genética , Feminino , Células Hep G2 , Humanos , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Pessoa de Meia-Idade , PTEN Fosfo-Hidrolase/metabolismo , Transdução de Sinais
18.
Anticancer Res ; 39(1): 191-200, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30591458

RESUMO

BACKGROUND/AIM: The present study assessed whether and how tumor cells undergoing hypoxia contribute to disease progression after moving to areas with different oxygen conditions. MATERIALS AND METHODS: Human colorectal carcinoma HCT116 cells cultured under mild hypoxia were subjected to in vivo experiments using transfer to immunodeficient murine recipients and to in vitro experiments using pharmacological inhibition of fatty acid ß-oxidation (FAO). RESULTS: Bone involvement and hepatic metastases were accelerated in transfer models of hypoxically cultured HCT116 cells. Hypoxic HCT116 cells exhibited FAO-dependent glycogen synthesis. FAO-dependent and -independent induction of gene expression also occurred under hypoxia. The distribution of glucose transporter 1 expression compared with heme oxygenase 1 expression in HCT116 cell spheroids seemed consistent with differential dependence of hypoxic expression of these molecules on FAO. CONCLUSION: These results provide insights into the contribution of hypoxia to tumor progression and the relevance of FAO.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Ácidos Graxos/antagonistas & inibidores , Transportador de Glucose Tipo 1/genética , Heme Oxigenase-1/genética , Animais , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Ácidos Graxos/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicogênio/biossíntese , Células HCT116 , Humanos , Camundongos , Oxirredução/efeitos dos fármacos , Oxigênio/metabolismo , Esferoides Celulares/efeitos dos fármacos , Hipóxia Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
19.
J Biol Chem ; 294(4): 1338-1348, 2019 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-30401744

RESUMO

The pathways for biosynthesis of glycogen in bacteria and starch in plants are evolutionarily and biochemically related. They are regulated primarily by ADP-glucose pyrophosphorylase, which evolved to satisfy metabolic requirements of a particular organism. Despite the importance of these two pathways, little is known about the mechanism that controls pyrophosphorylase activity or the location of its allosteric sites. Here, we report pyruvate-bound crystal structures of ADP-glucose pyrophosphorylase from the bacterium Agrobacterium tumefaciens, identifying a previously elusive activator site for the enzyme. We found that the tetrameric enzyme binds two molecules of pyruvate in a planar conformation. Each binding site is located in a crevice between the C-terminal domains of two subunits where they stack via a distinct ß-helix region. Pyruvate interacts with the side chain of Lys-43 and with the peptide backbone of Ser-328 and Gly-329 from both subunits. These structural insights led to the design of two variants with altered regulatory properties. In one variant (K43A), the allosteric effect was absent, whereas in the other (G329D), the introduced Asp mimicked the presence of pyruvate. The latter generated an enzyme that was preactivated and insensitive to further activation by pyruvate. Our study furnishes a deeper understanding of how glycogen biosynthesis is regulated in bacteria and the mechanism by which transgenic plants increased their starch production. These insights will facilitate rational approaches to enzyme engineering for starch production in crops of agricultural interest and will promote further study of allosteric signal transmission and molecular evolution in this important enzyme family.


Assuntos
Agrobacterium tumefaciens/enzimologia , Glucose-1-Fosfato Adenililtransferase/química , Glucose-1-Fosfato Adenililtransferase/metabolismo , Piruvatos/metabolismo , Sítios de Ligação , Glucose-1-Fosfato Adenililtransferase/genética , Glicogênio/biossíntese , Glicogênio/química , Modelos Moleculares , Estrutura Molecular
20.
J Cell Physiol ; 234(6): 9283-9296, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30317614

RESUMO

Hepatic stellate cells (HSCs) play an important microenvironmental role in hepatic progenitor cells (HPCs) differentiation fate. To reveal the specific mechanism of HSCs induced by transforming growth factor ß1 (TGF-ß1) signaling in HPCs differentiation process, we used Knockin and knockdown technologies induced by lentivirus to upregulate or downregulate TGF-ß1 level in mouse HSCs (mHSCs) (mHSCs-TGF-ß1 or mHSCs-TGF-ßR1sih3). Primary mouse HPCs (mHPCs) were isolated and were cocultured with mHSCs-TGF-ß1 and mHSCs-TGF-ßR1sih3 for 7 days. Differentiation of mHPCs was detected by quantitative reverse transcriptase polymerase chain reaction analysis and immunofluorence in vitro. mHPCs-E14.5 cell lines and differently treated mHSCs were cotransplanted into mice spleens immediately after establishment of acute liver injury model for animal studies. Engraftment and differentiation of transplanted cells as well as liver function recovery were measured at the seventh day via different methods. mHSCs-TGF-ß1 were transformed into myofibroblasts and highly expressed Jagged1, but that expression was reduced after blockage of TGF-ß1 signaling. mHPCs highly expressed downstream markers of Jagged1/Notch signaling and cholangiocyte markers (CK19, SOX9, and Hes1) after coculture with mHSCs-TGF-ß1 in vitro. In contrast, mature hepatocyte marker (ALB) was upregulated in mHPCs in coculture conditions with mHSCs-TGF-ßR1sih3. At the seventh day of cell transplantation assay, mHPCs-E 14.5 engrafted and differentiated into cholangiocytes after cotransplanting with TGF-ß1-knockin mHSCs, but the cells had a tendency to differentiate into hepatocytes when transplanted with TGF-ßR1-knockdown mHSCs, which corresponded to in vitro studies. HSCs play an important role in regulating HPCs differentiation into cholangiocytes via the TGF-ß1/Jagged1 signaling axis. However, HPCs have a tendency to differentiate into hepatocytes after blockage of TGF-ß1 signaling in HSCs.


Assuntos
Diferenciação Celular , Células Estreladas do Fígado/metabolismo , Proteína Jagged-1/metabolismo , Fígado/citologia , Transdução de Sinais , Células-Tronco/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Transdiferenciação Celular , Glicogênio/biossíntese , Hepatócitos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Miofibroblastos/metabolismo , Baço/transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA