Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 351
Filtrar
1.
Reprod Domest Anim ; 59(6): e14643, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38877774

RESUMEN

Progesterone has been shown to stimulate glycogen catabolism in uterine epithelial cells. Acid α-glucosidase (GAA) is an enzyme that breaks down glycogen within lysosomes. We hypothesized that progesterone may stimulate glycogenolysis in the uterine epithelium via GAA. We found that GAA was more highly expressed in the stroma on Day 1 than on Day 11. However, GAA did not appear to differ in the epithelium on Days 1 and 11. Progesterone (0-10 µM) had no effect on the levels of the full-length inactive protein (110 kDa) or the cleaved (active) peptides present inside the lysosome (70 and 76 kDa) in immortalized bovine uterine epithelial (BUTE) cells. Furthermore, the activity of GAA did not differ between the BUTE cells treated with 10 µM progesterone or control. Overall, we confirmed that GAA is present in the cow endometrium and BUTE cells. However, progesterone did not affect protein levels or enzyme activity.


Asunto(s)
Endometrio , Progesterona , alfa-Glucosidasas , Animales , Bovinos , Femenino , Endometrio/metabolismo , Endometrio/enzimología , Progesterona/farmacología , Progesterona/metabolismo , alfa-Glucosidasas/metabolismo , alfa-Glucosidasas/genética , Células Epiteliales/metabolismo , Glucogenólisis , Lisosomas/enzimología , Lisosomas/metabolismo , Glucógeno/metabolismo
2.
Sci Rep ; 14(1): 13670, 2024 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-38871968

RESUMEN

Cervical cancer, one of the most common gynecological cancers, is primarily caused by human papillomavirus (HPV) infection. The development of resistance to chemotherapy is a significant hurdle in treatment. In this study, we investigated the mechanisms underlying chemoresistance in cervical cancer by focusing on the roles of glycogen metabolism and the pentose phosphate pathway (PPP). We employed the cervical cancer cell lines HCC94 and CaSki by manipulating the expression of key enzymes PCK1, PYGL, and GYS1, which are involved in glycogen metabolism, through siRNA transfection. Our analysis included measuring glycogen levels, intermediates of PPP, NADPH/NADP+ ratio, and the ability of cells to clear reactive oxygen species (ROS) using biochemical assays and liquid chromatography-mass spectrometry (LC-MS). Furthermore, we assessed chemoresistance by evaluating cell viability and tumor growth in NSG mice. Our findings revealed that in drug-resistant tumor stem cells, the enzyme PCK1 enhances the phosphorylation of PYGL, leading to increased glycogen breakdown. This process shifts glucose metabolism towards PPP, generating NADPH. This, in turn, facilitates ROS clearance, promotes cell survival, and contributes to the development of chemoresistance. These insights suggest that targeting aberrant glycogen metabolism or PPP could be a promising strategy for overcoming chemoresistance in cervical cancer. Understanding these molecular mechanisms opens new avenues for the development of more effective treatments for this challenging malignancy.


Asunto(s)
Resistencia a Antineoplásicos , Glucógeno , Células Madre Neoplásicas , Fosfoenolpiruvato Carboxiquinasa (GTP) , Especies Reactivas de Oxígeno , Neoplasias del Cuello Uterino , Humanos , Femenino , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/patología , Especies Reactivas de Oxígeno/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Animales , Ratones , Línea Celular Tumoral , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Glucógeno/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Glucogenólisis , Vía de Pentosa Fosfato/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos
3.
Nat Metab ; 5(12): 2086-2093, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38066113

RESUMEN

Cotadutide is a dual glucagon-like peptide 1 and glucagon receptor agonist under development for the treatment of non-alcoholic steatohepatitis and type 2 diabetes mellitus (T2DM) and chronic kidney disease. Non-alcoholic steatohepatitis is a complex disease with no approved pharmacotherapies, arising from an underlying state of systemic metabolic dysfunction in association with T2DM and obesity. Cotadutide has been shown to improve glycaemic control, body weight, lipids, liver fat, inflammation and fibrosis. We conducted a two-part, randomized phase 2a trial in men and women with overweight or obesity diagnosed with T2DM to evaluate the efficacy and safety of cotadutide compared with placebo and liraglutide. The primary endpoints were change from baseline to day 28 of treatment in postprandial hepatic glycogen (part A) and to day 35 of treatment in fasting hepatic glycogen (part B) with cotadutide versus placebo. Secondary endpoints in part B were changes in fasting hepatic glycogen with cotadutide versus the mono glucagon-like peptide 1 receptor agonist, liraglutide, and change in hepatic fat fraction. The trial met its primary endpoint. We showed that cotadutide promotes greater reductions in liver glycogen and fat compared with placebo and liraglutide. Safety and tolerability findings with cotadutide were comparable to those of previous reports. Thus, this work provides evidence of additional benefits of cotadutide that could be attributed to glucagon receptor engagement. Our results suggest that cotadutide acts on the glucagon receptor in the human liver to promote glycogenolysis and improve the metabolic health of the liver. ClinicalTrials.gov registration: NCT03555994 .


Asunto(s)
Diabetes Mellitus Tipo 2 , Glucogenólisis , Enfermedad del Hígado Graso no Alcohólico , Masculino , Humanos , Femenino , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Sobrepeso/complicaciones , Sobrepeso/tratamiento farmacológico , Liraglutida/efectos adversos , Receptores de Glucagón/uso terapéutico , Glucógeno Hepático , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Péptidos/uso terapéutico , Enfermedad del Hígado Graso no Alcohólico/complicaciones
4.
J Clin Invest ; 133(24)2023 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-38099490

RESUMEN

Elevation of reactive oxygen species (ROS) levels is a general consequence of tumor cells' response to treatment and may cause tumor cell death. Mechanisms by which tumor cells clear fatal ROS, thereby rescuing redox balance and entering a chemoresistant state, remain unclear. Here, we show that cysteine sulfenylation by ROS confers on aryl hydrocarbon receptor (AHR) the ability to dissociate from the heat shock protein 90 complex but to bind to the PPP1R3 family member PPP1R3C of the glycogen complex in drug-treated tumor cells, thus activating glycogen phosphorylase to initiate glycogenolysis and the subsequent pentose phosphate pathway, leading to NADPH production for ROS clearance and chemoresistance formation. We found that basic ROS levels were higher in chemoresistant cells than in chemosensitive cells, guaranteeing the rapid induction of AHR sulfenylation for the clearance of excess ROS. These findings reveal that AHR can act as an ROS sensor to mediate chemoresistance, thus providing a potential strategy to reverse chemoresistance in patients with cancer.


Asunto(s)
Glucogenólisis , Neoplasias , Humanos , Especies Reactivas de Oxígeno/metabolismo , Resistencia a Antineoplásicos , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética
5.
Cell Death Dis ; 14(10): 664, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37816729

RESUMEN

Metabolic reprogramming has been recognized as one of the major mechanisms that fuel tumor initiation and progression. Our previous studies demonstrate that activation of Drp1 promotes fatty acid oxidation and downstream Wnt signaling. Here we investigate the role of Drp1 in regulating glycogen metabolism in colon cancer. Knockdown of Drp1 decreases mitochondrial respiration without increasing glycolysis. Analysis of cellular metabolites reveals that the levels of glucose-6-phosphate, a precursor for glycogenesis, are significantly elevated whereas pyruvate and other TCA cycle metabolites remain unchanged in Drp1 knockdown cells. Additionally, silencing Drp1 activates AMPK to stimulate the expression glycogen synthase 1 (GYS1) mRNA and promote glycogen storage. Using 3D organoids from Apcf/f/Villin-CreERT2 models, we show that glycogen levels are elevated in tumor organoids upon genetic deletion of Drp1. Similarly, increased GYS1 expression and glycogen accumulation are detected in xenograft tumors derived from Drp1 knockdown colon cancer cells. Functionally, increased glycogen storage provides survival advantage to Drp1 knockdown cells. Co-targeting glycogen phosphorylase-mediated glycogenolysis sensitizes Drp1 knockdown cells to chemotherapy drug treatment. Taken together, our results suggest that Drp1-loss activates glucose uptake and glycogenesis as compensative metabolic pathways to promote cell survival. Combined inhibition of glycogen metabolism may enhance the efficacy of chemotherapeutic agents for colon cancer treatment.


Asunto(s)
Neoplasias del Colon , Glucogenólisis , Humanos , Supervivencia Celular , Dinámicas Mitocondriales , Transformación Celular Neoplásica , Glucógeno/metabolismo , Neoplasias del Colon/genética , Dinaminas/metabolismo
6.
PLoS One ; 18(8): e0290832, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37651450

RESUMEN

Barth syndrome (BTHS) is an X-linked recessive genetic disorder due to mutations in the Tafazzin (TAFAZZIN) gene that lead to cardiac and skeletal muscle mitochondrial dysfunction. Previous studies in humans with BTHS demonstrate that the defects in muscle mitochondrial oxidative metabolism result in an enhanced reliance on anaerobic metabolism during exercise to meet energy demands of muscular work. During exercise, the liver normally increases glucose production via glycogenolysis and gluconeogenesis to match the elevated rate of muscle glucose uptake and meet the ATP requirements of working muscle. However, the impact of Tafazzin deficiency on hepatic glucose production and the pathways contributing to hepatic glucose production during exercise is unknown. Therefore, the purpose of this study was to quantify in vivo liver gluconeogenesis and glycogenolysis in Tafazzin knockdown mice at rest and during acute exercise. METHODS: Male TAFAZZIN shRNA transgenic (TG) and wild-type (WT) mice completed exhaustive treadmill running protocols to test exercise tolerance. Mice underwent 2H- and 13C-stable isotope infusions at rest and during a 30-minute treadmill running bout to quantify hepatic glucose production and associated nutrient fluxes under sedentary conditions and during acute exercise. Circulating and tissue (skeletal muscle and liver) samples were obtained during and following exercise to assess static metabolite levels. RESULTS: TG mice reached exhaustion sooner during exhaustive treadmill running protocols and exhibited higher plasma lactate concentrations after exhaustive exercise compared to WT mice. Arterial glucose levels were comparable between genotypes at rest, but higher in TG mice compared to WT mice during exercise. Consistent with the higher blood glucose, TG mice showed increased endogenous glucose production owing to elevated glycogenolysis compared to WT mice during exercise. Total gluconeogenesis, gluconeogenesis from glycerol, gluconeogenesis from phosphoenolpyruvate, pyruvate cycling, total cataplerosis, and anaplerotic fluxes were similar between TG and WT mice at rest and during exercise. However, lactate dehydrogenase flux and TCA cycle fluxes trended higher in TG mice during exercise. Liver glycogen content in TG was higher in TG vs. controls. CONCLUSION: Our data in the Tafazzin knockdown mouse suggest that elevated anaerobic metabolism during rest and exercise previously reported in humans with BTHS are supported by the finding of higher hepatic glycogenolysis.


Asunto(s)
Síndrome de Barth , Enfermedades Genéticas Ligadas al Cromosoma X , Glucogenólisis , Hiperglucemia , Humanos , Masculino , Animales , Ratones , Glucemia , Síndrome de Barth/genética , Hígado , Glucosa , Ratones Transgénicos , Músculo Esquelético
7.
Proc Natl Acad Sci U S A ; 120(25): e2300566120, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-37307453

RESUMEN

Mucosal-associated invariant T (MAIT) cells are a subset of unconventional T cells which recognize a limited repertoire of ligands presented by the MHC class-I like molecule MR1. In addition to their key role in host protection against bacterial and viral pathogens, MAIT cells are emerging as potent anti-cancer effectors. With their abundance in human, unrestricted properties, and rapid effector functions MAIT cells are emerging as attractive candidates for immunotherapy. In the current study, we demonstrate that MAIT cells are potent cytotoxic cells, rapidly degranulating and inducing target cell death. Previous work from our group and others has highlighted glucose metabolism as a critical process for MAIT cell cytokine responses at 18 h. However, the metabolic processes supporting rapid MAIT cell cytotoxic responses are currently unknown. Here, we show that glucose metabolism is dispensable for both MAIT cell cytotoxicity and early (<3 h) cytokine production, as is oxidative phosphorylation. We show that MAIT cells have the machinery required to make (GYS-1) and metabolize (PYGB) glycogen and further demonstrate that that MAIT cell cytotoxicity and rapid cytokine responses are dependent on glycogen metabolism. In summary, we show that glycogen-fueled metabolism supports rapid MAIT cell effector functions (cytotoxicity and cytokine production) which may have implications for their use as an immunotherapeutic agent.


Asunto(s)
Glucogenólisis , Células T Invariantes Asociadas a Mucosa , Humanos , Citocinas , Glucógeno , Glucosa
8.
Platelets ; 34(1): 2222184, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37292023

RESUMEN

Although the presence of glycogen in platelets was established in the 1960s, its importance to specific functions (i.e., activation, secretion, aggregation, and clot contraction) remains unclear. Patients with glycogen storage disease often present with increased bleeding and glycogen phosphorylase (GP) inhibitors, when used as treatments for diabetes, induce bleeding in preclinical studies suggesting some role for this form of glucose in hemostasis. In the present work, we examined how glycogen mobilization affects platelet function using GP inhibitors (CP316819 and CP91149) and a battery of ex vivo assays. Blocking GP activity increased glycogen levels in resting and thrombin-activated platelets and inhibited platelet secretion and clot contraction, with minimal effects on aggregation. Seahorse energy flux analysis and metabolite supplementation experiments suggested that glycogen is an important metabolic fuel whose role is affected by platelet activation and the availability of external glucose and other metabolic fuels. Our data shed light on the bleeding diathesis in glycogen storage disease patients and offer insights into the potential effects of hyperglycemia on platelets.


What did we know? Activated platelets transition from a low-energy-requiring, resting state to a high-energy-demanding state.Platelet glycogen is degraded upon activation.Glycogen storage disorders and glycogen phosphorylase inhibitors are associated with bleeding.What did we discover? Glycogen turnover occurs in resting platelets and its degradation is important for platelet functions.Glycogen phosphorylase inhibitors block secretion and clot contraction of which the latter can be reversed with alternative metabolic fuels.Glucose derived from glycogen may be routed through TCA/OxPhos versus aerobic glycolysis.What is the impact? Glycogen breakdown contributes to the high energy requirements of platelet function.Our work offers insights into potential energy sources in activated platelets.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno , Glucogenólisis , Trombosis , Humanos , Plaquetas/metabolismo , Glucosa/metabolismo , Glucosa/farmacología , Glucógeno/metabolismo , Glucógeno/farmacología , Trombosis/metabolismo , Enfermedad del Almacenamiento de Glucógeno/metabolismo
9.
J Physiol ; 601(12): 2293-2306, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37126218

RESUMEN

The precise regulation of blood glucose levels is indispensable for maintaining physiological functions. C1 neurons determine the outflow of the autonomic nervous and endocrine systems to maintain blood glucose levels in the body. In contrast, activation of C1 neurons induces a decrease in activity, suggesting that hypoactivity also participates in maintaining blood glucose levels. To examine this, we evaluated both glycogenolysis and hypometabolism induced by the selective activation of C1 neurons. We used DbhCre/0 mice expressing receptors for chemogenetic tools in C1 neurons, resulting from microinjection of the viral vector. C1 neurons were activated by intraperitoneal injection of clozapine N-oxide (CNO). The chemogenetic activation of C1 neurons significantly decreased body temperature, oxygen consumption and carbon dioxide production. On the other hand, blood glucose levels were increased by activation of C1 neurons 2 h after CNO administration, even in the fasting state. In this situation, an increase in glucagon and corticosterone levels was observed, while hepatic glycogen content decreased significantly. Plasma insulin levels were not changed by the activation of C1 neurons despite the increase in blood glucose level. Furthermore, adrenal sympathetic nerve activity was significantly increased by the activation of C1 neurons, and plasma catecholamine levels increased significantly. In conclusion, the selective activation of C1 neurons using chemogenetic tools induced an increase in blood glucose levels, probably as a result of hepatic glycogenolysis and hypometabolism. KEY POINTS: Chemogenetic activation of C1 neurons in medulla oblongata decreased body temperature. Oxygen consumption and carbon dioxide production were decreased by chemogenetic activation of C1 neurons in medulla oblongata. Blood glucose levels were increased by chemogenetic activation of C1 neurons in medulla oblongata. Chemogenetic activation of C1 neurons in medulla oblongata increased glucagon, corticosterone and catecholamine levels in plasma. An increase in blood glucose levels by activation of C1 neurons occurred due to the combined effect of hepatic glycogenolysis and hypometabolism.


Asunto(s)
Glucemia , Glucogenólisis , Ratones , Animales , Glucagón , Corticosterona/farmacología , Dióxido de Carbono , Neuronas/fisiología , Bulbo Raquídeo/fisiología , Catecolaminas
10.
Cell Biochem Funct ; 41(5): 609-618, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37255029

RESUMEN

Interleukin 6 (IL6) is an multifunctional cytokine that modulates several biological responses, including glucose metabolism. However, its acute effects on hepatic glucose release are still uncertain. The main purpose of this study was to investigate the effects of IL6 on gluconeogenesis from several glucose precursors (alanine, pyruvate and glutamine) and on the suppressive action of insulin on cAMP-stimulated glycogen catabolism in rat liver. IL6 effect on insulin peripheral sensitivity was also evaluated. IL6 was injected intravenously into rats and, 1 h later, gluconeogenesis and glycogenolysis were assessed in liver perfusion and peripheral insulin sensitivity by insulin tolerance test (ITT). IL6 intravenous injection increased hepatic glucose production from alanine, without changing pyruvate, lactate and urea production. IL6 injection also increased hepatic glucose production from pyruvate and glutamine. In addition, IL6 decreased the suppressive effect of insulin on cAMP-stimulated glucose and lactate production and glycogenolysis, without affecting pyruvate production. Furthermore, IL6 reduced the plasma glucose disappearance constant (kITT), an indicator of insulin resistance. In conclusion, IL6 acutely increased hepatic glucose release (gluconeogenesis and glycogenolysis) by a mechanism that likely involved the induction of insulin resistance in the liver, as evidenced by the reduced suppressive effect of insulin on cAMP-stimulated glycogen catabolism. In consistency, IL6 acutely induced peripheral insulin resistance.


Asunto(s)
Glucogenólisis , Resistencia a la Insulina , Ratas , Animales , Gluconeogénesis , Insulina/farmacología , Insulina/metabolismo , Interleucina-6/metabolismo , Glutamina/metabolismo , Glutamina/farmacología , Glucosa/farmacología , Glucosa/metabolismo , Glucógeno/metabolismo , Glucógeno/farmacología , Hígado/metabolismo , Ácido Láctico/farmacología , Ácido Láctico/metabolismo , Piruvatos/metabolismo , Piruvatos/farmacología , Alanina/farmacología , Alanina/metabolismo , Glucemia
11.
Carbohydr Polym ; 299: 120200, 2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-36876811

RESUMEN

It has been reported that glycogen in Escherichia coli has two structural states, that is, fragility and stability, which alters dynamically. However, molecular mechanisms behind the structural alterations are not fully understood. In this study, we focused on the potential roles of two important glycogen degradation enzymes, glycogen phosphorylase (glgP) and glycogen debranching enzyme (glgX), in glycogen structural alterations. The fine molecular structure of glycogen particles in Escherichia coli and three mutants (ΔglgP, ΔglgX and ΔglgP/ΔglgX) were examined, which showed that glycogen in E. coli ΔglgP and E. coli ΔglgP/ΔglgX were consistently fragile while being consistently stable in E. coli ΔglgX, indicating the dominant role of GP in glycogen structural stability control. In sum, our study concludes that glycogen phosphorylase is essential in glycogen structural stability, leading to molecular insights into structural assembly of glycogen particles in E. coli.


Asunto(s)
Sistema de la Enzima Desramificadora del Glucógeno , Glucogenólisis , Escherichia coli , Citoplasma , Glucógeno
12.
J Genet ; 1022023.
Artículo en Inglés | MEDLINE | ID: mdl-36814107

RESUMEN

Duchenne muscular dystrophy (DMD) is the most common form of progressive childhood muscular dystrophy associated with weakness of limbs, loss of ambulation, heart weakness and early death. The mutations causing either loss-of-expression or function of the full-length protein dystrophin (Dp427) from the DMD gene are responsible for the disease pathology. Dp427 forms a part of the large dystroglycan complex, called DAPC, in the sarcolemma, and its absence derails muscle contraction. Muscle biopsies from DMD patients show an overactivation of excitation-contraction-coupling (ECC) activable calcium incursion, sarcolemmal ROS production, NHE1 activation, IL6 secretion, etc. The signalling pathways, like Akt/PBK, STAT3, p38MAPK, and ERK1/2, are also hyperactive in DMD. These pathways are responsible for post-mitotic trophic growth and metabolic adaptation, in response to exercise in healthy muscles, but cause atrophy and cell death in dystrophic muscles. We hypothesize that the metabolic background of repressed glycolysis in DMD, as opposed to excess glycolysis seen in cancers or healthy contracting muscles, changes the outcome of these 'growth pathways'. The reduced glycolysis has been considered a secondary outcome of the cytoskeletal disruptions seen in DMD. Given the cytoskeleton-crosslinking ability of the glycolytic enzymes, we hypothesize that the failure of glycogenolytic and glycolytic enzymes to congregate is the primary pathology, which then affects the subsarcolemmal cytoskeletal organization in costameres and initiates the pathophysiology associated with DMD, giving rise to the tissue-specific differences in disease progression between muscle, heart and brain. The lacunae in the regulation of the key components of the hypothesized metabolome, and the limitations of this theory are deliberated. The considerations for developing future therapies based on known pathological processes are also discussed.


Asunto(s)
Glucogenólisis , Distrofia Muscular de Duchenne , Humanos , Niño , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Costameras/metabolismo , Costameras/patología , Distrofina/genética , Distrofina/metabolismo , Músculos/metabolismo , Músculos/patología , Sarcolema/metabolismo , Sarcolema/patología , Músculo Esquelético/metabolismo
13.
Am J Physiol Endocrinol Metab ; 324(2): E199-E208, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36652399

RESUMEN

It has been proposed that brain glucagon action inhibits glucagon-stimulated hepatic glucose production (HGP), which may explain, at least in part, why glucagon's effect on HGP is transient. However, the pharmacologic off-target effects of glucagon in the brain may have been responsible for previously observed effects. Therefore, the aim of this study was to determine if central glucagon action plays a physiologic role in the regulation of HGP. Insulin was maintained at baseline while glucagon was either infused into the carotid and vertebral arteries or into a peripheral (leg) vein at rates designed to increase glucagon in the head in one group, while keeping glucagon at the liver matched between groups. The extraction rate of glucagon across the head was high (double that of the liver), and hypothalamic cAMP increased twofold, in proportion to the exposure of the brain to increased glucagon, but HGP was not reduced by the increase in brain glucagon signaling, as had been suggested previously (the areas under the curve for HGP were 840 ± 14 vs. 871 ± 36 mg/kg/240 min in head vs. peripheral infusion groups, respectively). Central nervous system glucagon action reduced circulating free fatty acids and glycerol, and this was associated with a modest reduction in net hepatic gluconeogenic flux. However, offsetting autoregulation by the liver (i.e., a reciprocal increase in net hepatic glycogenolysis) prevented a change in HGP. Thus, while physiologic engagement of the brain by glucagon can alter hepatic carbon flux, it does not appear to be responsible for the transient fall in HGP that occurs following the stimulation of HGP during a square wave rise in glucagon.NEW & NOTEWORTHY Glucagon stimulates hepatic glucose production through its direct effects on the liver but may indirectly inhibit this process by acting on the brain. This was tested by delivering glucagon via the cerebral circulatory system. Central nervous system glucagon action reduced liver gluconeogenic flux, but glycogenolysis increased, resulting in no net change in hepatic glucose production. Surprisingly, brain glucagon also appeared to suppress lipolysis (plasma free fatty acid and glycerol levels were reduced).


Asunto(s)
Glucagón , Glucogenólisis , Glucemia/metabolismo , Encéfalo/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Glucagón/metabolismo , Gluconeogénesis , Glucosa/metabolismo , Glicerol/metabolismo , Insulina/metabolismo , Hígado/metabolismo , Animales
14.
FEBS Lett ; 596(22): 2940-2951, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36050761

RESUMEN

Motor neurons in the spinal cord are essential for movement. During the embryonic period, developing motor neurons store glycogen to protect against hypoglycemic and hypoxic stress. However, the mechanisms by which glycogen metabolism is regulated in motor neurons remain unclear. We herein investigated the transcriptional regulation of genes related to glycogen metabolism in the developing spinal cord. We focused on the regulatory mechanism of glycogen synthase (Gys1) and glycogen phosphorylase brain isoform (PygB), which play central roles in glycogen metabolism, and found that the transcription factor STAT3 regulated the expression of Gys1 and PygB via cis-regulatory promoter sequences in the developing spinal cord. These results suggest that STAT3 is important for the regulation of glycogen metabolism during motor neuron development.


Asunto(s)
Glucógeno Sintasa , Glucogenólisis , Glucógeno Sintasa/genética , Glucógeno Sintasa/metabolismo , Glucógeno/metabolismo , Neuronas Motoras/metabolismo , Regulación de la Expresión Génica
15.
Int J Mol Sci ; 23(17)2022 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-36077341

RESUMEN

Glycogen storage disease type IX (GSD-IX) constitutes nearly a quarter of all GSDs. This ketotic form of GSD is caused by mutations in phosphorylase kinase (PhK), which is composed of four subunits (α, ß, γ, δ). PhK is required for the activation of the liver isoform of glycogen phosphorylase (PYGL), which generates free glucose-1-phosphate monomers to be used as energy via cleavage of the α -(1,4) glycosidic linkages in glycogen chains. Mutations in any of the PhK subunits can negatively affect the regulatory and catalytic activity of PhK during glycogenolysis. To understand the pathogenesis of GSD-IX-beta, we characterized a newly created PHKB knockout (Phkb−/−) mouse model. In this study, we assessed fasting blood glucose and ketone levels, serum metabolite concentrations, glycogen phosphorylase activity, and gene expression of gluconeogenic genes and fibrotic genes. Phkb−/− mice displayed hepatomegaly with lower fasting blood glucose concentrations. Phkb−/− mice showed partial liver glycogen phosphorylase activity and increased sensitivity to pyruvate, indicative of partial glycogenolytic activity and upregulation of gluconeogenesis. Additionally, gene expression analysis demonstrated increased lipid metabolism in Phkb−/− mice. Gene expression analysis and liver histology in the livers of old Phkb−/− mice (>40 weeks) showed minimal profibrogenic features when analyzed with age-matched wild-type (WT) mice. Collectively, the Phkb−/− mouse recapitulates mild clinical features in patients with GSD-IX-beta. Metabolic and molecular analysis confirmed that Phkb−/− mice were capable of sustaining energy homeostasis during prolonged fasting by using partial glycogenolysis, increased gluconeogenesis, and potentially fatty acid oxidation in the liver.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno , Glucogenólisis , Fosforilasa Quinasa/metabolismo , Animales , Glucemia/metabolismo , Modelos Animales de Enfermedad , Enfermedad del Almacenamiento de Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno/metabolismo , Hígado/metabolismo , Ratones , Fosforilasa Quinasa/genética
16.
J Sep Sci ; 45(20): 3791-3799, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35964279

RESUMEN

5-Isopropyl-4-(2-chlorophenyl)-1-ethyl-1,4-dihydro-6-methyl-2,3,5-pyridinetricarboxylic acid ester disodium salt hydrate, is a noncompetitive inhibitor of glycogen phosphorylase - a critical enzyme in the process of glycogenolysis. This chemical compound is most widely used in studies focused on the inhibition of liver and muscle glycogenolysis. However, there are also reports linking phosphorylase inhibitor action with cognitive function and glycogen metabolism in the brain. The aim of this study was to develop and validate the liquid chromatography-mass spectrometry method for quantitative analysis of present chemical compound in mouse tissues including different brain regions. Obtained linearity was in the range of 10-550 ng/mL with a correlation coefficient of 0.9996. In tissue matrix samples the limit of detection was 7.76 ng/mL, while the limit of quantification was 23.29 ng/mL. The coefficient of variation values did not exceed ±15% for either within a run or between run precision quality control samples. The extraction recovery was between 89.44% and 98.70% for various validation analyte concentrations. The present method was successful in the quantitative determination of the presented analyte in mouse tissues and provided evidence that the compound is not only present in the liver, heart, and skeletal muscle but also in different regions of brain tissue such as the hippocampus, cerebellum, and cortex.


Asunto(s)
Glucogenólisis , Animales , Ratones , Ésteres , Cromatografía Liquida , Espectrometría de Masas , Fosforilasas , Músculo Esquelético , Cromatografía Líquida de Alta Presión/métodos , Reproducibilidad de los Resultados
17.
Cell Rep ; 40(1): 111041, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35793618

RESUMEN

Glycogen is the primary energy reserve in mammals, and dysregulation of glycogen metabolism can result in glycogen storage diseases (GSDs). In muscle, glycogen synthesis is initiated by the enzymes glycogenin-1 (GYG1), which seeds the molecule by autoglucosylation, and glycogen synthase-1 (GYS1), which extends the glycogen chain. Although both enzymes are required for proper glycogen production, the nature of their interaction has been enigmatic. Here, we present the human GYS1:GYG1 complex in multiple conformations representing different functional states. We observe an asymmetric conformation of GYS1 that exposes an interface for close GYG1 association, and propose this state facilitates handoff of the GYG1-associated glycogen chain to a GYS1 subunit for elongation. Full activation of GYS1 widens the GYG1-binding groove, enabling GYG1 release concomitant with glycogen chain growth. This structural mechanism connecting chain nucleation and extension explains the apparent stepwise nature of glycogen synthesis and suggests distinct states to target for GSD-modifying therapeutics.


Asunto(s)
Glucógeno Sintasa , Glucogenólisis , Glicoproteínas , Glucosiltransferasas/metabolismo , Glucógeno/metabolismo , Glucógeno Sintasa/metabolismo , Glicoproteínas/metabolismo , Humanos
18.
Toxicol Lett ; 366: 72-80, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35817173

RESUMEN

Decabromodiphenyl ethane (DBDPE) as a novel brominated flame retardant is frequently detected in environmental media due to its widespread use. Studies have shown that exposure to environmental pollutants in utero could lead to weight loss in newborns and obesity in adulthood. However, the mechanisms of how the cubs grow rapidly from low birth weight to obesity remain unclear. Although it has been reported that perinatal DBDPE exposure caused obesity in the offspring of mice in adulthood, its metabolic changes in offspring juvenile are unknown. Here, we monitored changes of body weight in cubs following exposure to DBDPE in utero. Furthermore, 1H NMR-based metabolomics was used to investigate the effects of in utero DBDPE exposure on the metabolic profile of neonates included days 1 and 25. Additionally, the expression levels of key genes in the relevant pathways were quantified. The results showed that exposure to high levels of DBDPE in utero caused cubs to be smaller at birth and grow rapidly during lactation. Metabolomics analysis showed that cubs in the exposed group consistently accelerated glycogenolysis and lipid synthesis to promote appetite and energy absorption, and achieved rapid growth in the subsequent lactation, which was further evidenced by the expression levels of genes related to glycolipid metabolism. These results reveal the mechanism for the first time how the weight loss of newborns in utero environmental pollutant exposure transformed into obesity in adulthood, although the exposures here were much higher than would be anticipated from the environment.


Asunto(s)
Contaminantes Ambientales , Retardadores de Llama , Glucogenólisis , Animales , Bromobencenos , Femenino , Retardadores de Llama/metabolismo , Retardadores de Llama/toxicidad , Éteres Difenilos Halogenados/toxicidad , Lípidos , Ratones , Obesidad , Embarazo , Pérdida de Peso
19.
J Biol Chem ; 298(7): 102093, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35654138

RESUMEN

Autophagy is an essential cellular process involving degradation of superfluous or defective macromolecules and organelles as a form of homeostatic recycling. Initially proposed to be a "bulk" degradation pathway, a more nuanced appreciation of selective autophagy pathways has developed in the literature in recent years. As a glycogen-selective autophagy process, "glycophagy" is emerging as a key metabolic route of transport and delivery of glycolytic fuel substrate. Study of glycophagy is at an early stage. Enhanced understanding of this major noncanonical pathway of glycogen flux will provide important opportunities for new insights into cellular energy metabolism. In addition, glycogen metabolic mishandling is centrally involved in the pathophysiology of several metabolic diseases in a wide range of tissues, including the liver, skeletal muscle, cardiac muscle, and brain. Thus, advances in this exciting new field are of broad multidisciplinary interest relevant to many cell types and metabolic states. Here, we review the current evidence of glycophagy involvement in homeostatic cellular metabolic processes and of molecular mediators participating in glycophagy flux. We integrate information from a variety of settings including cell lines, primary cell culture systems, ex vivo tissue preparations, genetic disease models, and clinical glycogen disease states.


Asunto(s)
Autofagia , Glucógeno , Glucogenólisis , Autofagia/fisiología , Glucógeno/metabolismo , Macroautofagia
20.
Mol Cell ; 82(16): 3077-3088.e6, 2022 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-35738262

RESUMEN

Glycolysis facilitates the rapid recall response of CD8+ memory T (Tm) cells. However, it remains unclear whether Tm cells uptake exogenous glucose or mobilize endogenous sugar to fuel glycolysis. Here, we show that intracellular glycogen rather than extracellular glucose acts as the major carbon source for the early recall response. Following antigenic stimulation, Tm cells exhibit high glycogen phosphorylase (brain form, PYGB) activity, leading to glycogenolysis and release of glucose-6-phosphate (G6P). Elevated G6P mainly flows to glycolysis but is also partially channeled to the pentose phosphate pathway, which maintains the antioxidant capacity necessary for later recall stages. Mechanistically, TCR signaling directly induces phosphorylation of PYGB by LCK-ZAP70. Functionally, the glycogenolysis-fueled early recall response of CD8+ Tm cells accelerates the clearance of OVA-Listeria monocytogenes in an infected mouse model. Thus, we uncover a specific dependency on glycogen for the initial activation of memory T cells, which may have therapeutic implications for adaptive immunity.


Asunto(s)
Glucogenólisis , Animales , Linfocitos T CD8-positivos , Glucosa/metabolismo , Glucógeno/metabolismo , Células T de Memoria , Ratones , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA