Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 38
Filtrer
1.
BMC Biotechnol ; 24(1): 58, 2024 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-39174975

RÉSUMÉ

Based on our previous findings that salicylic acid and jasmonic acid increased Nostoc flagelliforme polysaccharide yield by regulating intracellular nitric oxide (NO) levels, the mechanism through which NO affects polysaccharide biosynthesis in Nostoc flagelliforme was explored from the perspective of S-nitrosylation (SNO). The addition of NO donor and scavenger showed that intracellular NO had a significant positive effect on the polysaccharide yield of N. flagelliforme. To explore the mechanism, we investigated the relationship between NO levels and the activity of several key enzymes involved in polysaccharide biosynthesis, including fructose 1,6-bisphosphate aldolase (FBA), glucokinase (GK), glucose 6-phosphate dehydrogenase (G6PDH), mitochondrial isocitrate dehydrogenase (ICDH), and UDP-glucose dehydrogenase (UGDH). The enzymatic activities of G6PDH, ICDH, and UGDH were shown to be significantly correlated with the shifts in intracellular NO levels. For further validation, G6PDH, ICDH, and UGDH were heterologously expressed in Escherichia coli and purified via Ni+-NAT affinity chromatography, and subjected to a biotin switch assay and western blot analysis, which revealed that UGDH and G6PDH were susceptible to SNO. Furthermore, mass spectrometry analysis of proteins treated with S-nitrosoglutathione (GSNO) identified the SNO modification sites for UGDH and G6PDH as cysteine 423 and cysteine 249, respectively. These findings suggest that NO modulates polysaccharide biosynthesis in N. flagelliforme through SNO of UGDH and G6PDH. This reveals a potential mechanism through which NO promotes polysaccharide synthesis in N. flagelliforme, while also providing a new strategy for improving the industrial production of polysaccharides.


Sujet(s)
Monoxyde d'azote , Nostoc , Nostoc/métabolisme , Nostoc/enzymologie , Nostoc/génétique , Monoxyde d'azote/métabolisme , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/génétique , Polyosides bactériens/métabolisme , Polyosides bactériens/biosynthèse , Polyosides/métabolisme , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Escherichia coli/génétique , Escherichia coli/métabolisme
3.
Immun Inflamm Dis ; 12(8): e1341, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39092715

RÉSUMÉ

BACKGROUND: Sirtuin 7 (SIRT7) is pivotal in diverse diseases progression. Importantly, SIRT7 is associated with melanin production. However, whether SIRT7 regulates vitiligo is unclear. Therefore, we aimed to investigate the effects of SIRT7 on pigmentation and the modification of glucose 6-phosphate dehydrogenase (G6PD). METHODS: After knockdown SIRT7 and G6PD, pigmentation of melanocytes was evaluated using commercial kits, immunofluorescence, and Western blot analysis. The succinylation of G6PD mediated by SIRT7 was analyzed using co-immunoprecipitation, immunofluorescence, Western blot analysis, and cycloheximide-chase experiment. RESULTS: We found that SIRT7 was highly expressed in vitiligo skin lesions. Knockdown of SIRT7 increased tyrosinase activity, melanin content, and the levels of α-melanocyte-stimulating hormone, MITF, TYR, TRP1, and TRP2. Additionally, SIRT7 directly interacted with G6PD. Silenced SIRT7 promoted the succinylation of G6PD and enhanced its protein stability. G6PD knockdown reversed the effect of reduced SIRT7 expression on melanin production. CONCLSUION: Silencing of SIRT7 promotes pigmentation of melanocytes by succinylating G6PD, suggesting that SIRT7-mediated G6PD desuccinylation may promote vitiligo progression.


Sujet(s)
Évolution de la maladie , Glucose 6-phosphate dehydrogenase , Mélanines , Mélanocytes , Sirtuines , Vitiligo , Vitiligo/métabolisme , Vitiligo/anatomopathologie , Humains , Mélanocytes/métabolisme , Mélanocytes/anatomopathologie , Sirtuines/métabolisme , Sirtuines/génétique , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/génétique , Mélanines/métabolisme , Mélanines/biosynthèse
4.
Cell Death Dis ; 15(7): 516, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39025830

RÉSUMÉ

Tumour metabolic reprogramming is pivotal for tumour survival and proliferation. Investigating potential molecular mechanisms within the heterogeneous and clinically aggressive triple-negative breast cancer (TNBC) subtype is essential to identifying novel therapeutic targets. Accordingly, we investigated the role of branched-chain α-keto acid dehydrogenase kinase (BCKDK) in promoting tumorigenesis in TNBC. We analysed The Cancer Genome Atlas dataset and immunohistochemically stained surgical specimens to investigate BCKDK expression and its prognostic implications in TNBC. The effects of BCKDK on tumorigenesis were assessed using cell viability, colony formation, apoptosis, and cell cycle assays, and subsequently validated in vivo. Metabolomic screening was performed via isotope tracer studies. The downstream target was confirmed using mass spectrometry and a co-immunoprecipitation experiment coupled with immunofluorescence analysis. Upstream transcription factors were also examined using chromatin immunoprecipitation and luciferase assays. BCKDK was upregulated in TNBC tumour tissues and associated with poor prognosis. BCKDK depletion led to reduced cell proliferation both in vitro and vivo. MYC-associated zinc finger protein (MAZ) was confirmed as the major transcription factor directly regulating BCKDK expression in TNBC. Mechanistically, BCKDK interacted with glucose-6-phosphate dehydrogenase (G6PD), leading to increased flux in the pentose phosphate pathway for macromolecule synthesis and detoxification of reactive oxygen species. Forced expression of G6PD rescued the growth defect in BCKDK-deficient cells. Notably, the small-molecule inhibitor of BCKDK, 3,6-dichlorobenzo(b)thiophene-2-carboxylic acid, exhibited anti-tumour effects in a patient-derived tumour xenograft model. Our findings hold significant promise for developing targeted therapies aimed at disrupting the MAZ/BCKDK/G6PD signalling pathway, offering potential advancements in treating TNBC through metabolic reprogramming.


Sujet(s)
Prolifération cellulaire , Glucose , Glucose 6-phosphate dehydrogenase , Tumeurs du sein triple-négatives , Régulation positive , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/génétique , Humains , Femelle , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/génétique , Animaux , Lignée cellulaire tumorale , Souris , Glucose/métabolisme , Facteurs de transcription/métabolisme , Régulation de l'expression des gènes tumoraux , Souris nude
5.
Int Immunopharmacol ; 139: 112688, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39029227

RÉSUMÉ

Chondrocytes, known for their metabolic adaptability in response to varying stimuli, play a significant role in osteoarthritis (OA) progression. Glucose-6-phosphate dehydrogenase (G6PD), the rate-limiting enzyme of the pentose phosphate pathway, has recently been found to upregulate in OA chondrocyte. However, the exact role of G6PD in temporomandibular joint osteoarthritis (TMJOA) and its effect on chondrocyte function remains unclear. In present study, we induced OA-like conditions in the rat temporomandibular joint via occlusal disharmony (OD), noting a marked increase in G6PD expression in the condylar cartilage. Our data show that G6PD knockdown in mandibular condylar chondrocytes (MCCs) reduces the expression of catabolic enzymes (e.g., MMP3, MMP13) and inflammatory cytokines (e.g., IL6) induced by IL-1ß. G6PD knockdown also mitigates IL-1ß-induced upregulation of ERK, JNK, and p38 phosphorylation and reduces reactive oxygen species (ROS) levels by decreasing the nicotinamide adenine dinucleotide phosphate (NADPH) and NADPH oxidases 4 (NOX4) mRNA expression. In summary, G6PD appears to regulate the inflammatory state of condylar chondrocytes via the NOX-ROS-MAPK axis, highlighting its potential as a therapeutic target for TMJOA.


Sujet(s)
Chondrocytes , Glucose 6-phosphate dehydrogenase , NADPH Oxidase 4 , Arthrose , Espèces réactives de l'oxygène , Animaux , NADPH Oxidase 4/métabolisme , NADPH Oxidase 4/génétique , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Arthrose/métabolisme , Espèces réactives de l'oxygène/métabolisme , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/génétique , Mâle , Rats , Rat Sprague-Dawley , Interleukine-1 bêta/métabolisme , Articulation temporomandibulaire/anatomopathologie , Articulation temporomandibulaire/métabolisme , Cellules cultivées , Cytokines/métabolisme , Cartilage articulaire/anatomopathologie , Cartilage articulaire/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Transduction du signal , Modèles animaux de maladie humaine , Humains
6.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167444, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39074627

RÉSUMÉ

The glucose-6-phosphate dehydrogenase (G6PD) deficiency is X-linked and is the most common enzymatic deficiency disorder globally. It is a crucial enzyme for the pentose phosphate pathway and produces NADPH, which plays a vital role in regulating the oxidative stress of many cell types. The deficiency of G6PD primarily causes hemolytic anemia under oxidative stress triggered by food, drugs, or infection. G6PD-deficient patients infected with SARS-CoV-2 showed an increase in hemolysis and thrombosis. Patients also exhibited prolonged COVID-19 symptoms, ventilation support, neurological impacts, and high mortality. However, the mechanism of COVID-19 severity in G6PD deficient patients and its neurological manifestation is still ambiguous. Here, using a CRISPR-edited G6PD deficient human microglia cell culture model, we observed a significant reduction in NADPH level and an increase in basal reactive oxygen species (ROS) in microglia. Interestingly, the deficiency of the G6PD-NAPDH axis impairs induced nitric oxide synthase (iNOS) mediated nitric oxide (NO) production, which plays a fundamental role in inhibiting viral replication. Surprisingly, we also observed that the deficiency of the G6PD-NADPH axis reduced lysosomal acidification and free radical production, further abrogating the lysosomal clearance of viral particles. Thus, impairment of NO production, lysosomal functions, and redox dysregulation in G6PD deficient microglia altered innate immune response, promoting the severity of SARS-CoV-2 pathogenesis.


Sujet(s)
COVID-19 , Déficit en glucose-6-phosphate-déshydrogénase , Lysosomes , Microglie , Nitric oxide synthase type II , Phagocytose , Humains , COVID-19/métabolisme , COVID-19/anatomopathologie , COVID-19/virologie , COVID-19/immunologie , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/génétique , Déficit en glucose-6-phosphate-déshydrogénase/métabolisme , Déficit en glucose-6-phosphate-déshydrogénase/anatomopathologie , Déficit en glucose-6-phosphate-déshydrogénase/génétique , Lysosomes/métabolisme , Microglie/métabolisme , Microglie/anatomopathologie , NADP/métabolisme , Monoxyde d'azote/métabolisme , Nitric oxide synthase type II/métabolisme , Nitric oxide synthase type II/génétique , Stress oxydatif , Espèces réactives de l'oxygène/métabolisme , Cellules cultivées
7.
Sci Rep ; 14(1): 16029, 2024 07 11.
Article de Anglais | MEDLINE | ID: mdl-38992151

RÉSUMÉ

Glucose-6-phosphate dehydrogenase (G6PD) deficiency is one of the most common enzymopathies worldwide. Patients with G6PD deficiency are usually asymptomatic throughout their life but can develop acute hemolysis after exposure to free radicals or certain medications. Several studies have shown that serum miRNAs can be used as prognostic biomarkers in various types of hemolytic anemias. However, the impact of G6PD deficiency on circulating miRNA profiles is largely unknown. The present study aimed to assess the use of serum miRNAs as biomarkers for detecting hemolysis in the nonacute phase of G6PD deficiency. Patients with severe or moderate G6PD Viangchan (871G > A) deficiency and normal G6PD patients were enrolled in the present study. The biochemical hemolysis indices were normal in the three groups, while the levels of serum miR-451a, miR-16, and miR-155 were significantly increased in patients with severe G6PD deficiency. In addition, 3D analysis of a set of three miRNAs (miR-451a, miR-16, and miR-155) was able to differentiate G6PD-deficient individuals from healthy individuals, suggesting that these three miRNAs may serve as potential biomarkers for patients in the nonhemolytic phase of G6PD deficiency. In conclusion, miRNAs can be utilized as additional biomarkers to detect hemolysis in the nonacute phase of G6PD deficiency.


Sujet(s)
Marqueurs biologiques , Déficit en glucose-6-phosphate-déshydrogénase , Hémolyse , microARN , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Marqueurs biologiques/sang , Études cas-témoins , Glucose 6-phosphate dehydrogenase/génétique , Glucose 6-phosphate dehydrogenase/sang , Déficit en glucose-6-phosphate-déshydrogénase/sang , Déficit en glucose-6-phosphate-déshydrogénase/diagnostic , Déficit en glucose-6-phosphate-déshydrogénase/génétique , microARN/sang
8.
Nat Commun ; 15(1): 5857, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997257

RÉSUMÉ

Cancer cells depend on nicotinamide adenine dinucleotide phosphate (NADPH) to combat oxidative stress and support reductive biosynthesis. One major NADPH production route is the oxidative pentose phosphate pathway (committed step: glucose-6-phosphate dehydrogenase, G6PD). Alternatives exist and can compensate in some tumors. Here, using genetically-engineered lung cancer mouse models, we show that G6PD ablation significantly suppresses KrasG12D/+;Lkb1-/- (KL) but not KrasG12D/+;P53-/- (KP) lung tumorigenesis. In vivo isotope tracing and metabolomics reveal that G6PD ablation significantly impairs NADPH generation, redox balance, and de novo lipogenesis in KL but not KP lung tumors. Mechanistically, in KL tumors, G6PD ablation activates p53, suppressing tumor growth. As tumors progress, G6PD-deficient KL tumors increase an alternative NADPH source from serine-driven one carbon metabolism, rendering associated tumor-derived cell lines sensitive to serine/glycine depletion. Thus, oncogenic driver mutations determine lung cancer dependence on G6PD, whose targeting is a potential therapeutic strategy for tumors harboring KRAS and LKB1 co-mutations.


Sujet(s)
Glucose 6-phosphate dehydrogenase , Homéostasie , Tumeurs du poumon , NADP , Oxydoréduction , Protein-Serine-Threonine Kinases , Protéines proto-oncogènes p21(ras) , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/génétique , Animaux , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , NADP/métabolisme , Souris , Humains , Lignée cellulaire tumorale , Lipogenèse/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , AMP-activated protein kinase kinases/génétique , AMP-activated protein kinase kinases/métabolisme , Voie des pentoses phosphates/génétique , AMP-Activated Protein Kinases/métabolisme , Mâle , Souris knockout , Femelle , Mutation
10.
Clin Lab ; 70(6)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38868868

RÉSUMÉ

BACKGROUND: Klinefelter syndrome is a common sex chromosome abnormality in males, characterized by an extra X chromosome compared to normal males. Glucose-6-phosphate dehydrogenase deficiency (G6PD) is an X-linked incomplete dominant defect disorder. In this study, we reported the unexpected detection of Klinefelter syndrome in a patient with G6PD. METHODS: G6PD enzyme activity was measured by immunoenzyme assay, and genetic analysis was performed using a fluorescent PCR melting curve method (PCR-melting curve). Sex chromosome number abnormalities were detected by multiplex ligation-dependent probe amplification (MLPA). The patient also underwent peripheral blood chromosome karyotype analysis. RESULTS: The patient's G6PD and 6PGD enzyme activities were 21.34 U/L and 22.85 U/L, respectively, and their ratio was below the reference range (0.93). The PCR-melting curve displayed a c.1388 heterozygous mutation in this boy, and the Sanger sequencing provided the same results. MLPA results suggested the presence of approxi-mately two copies of the X-chromosome in the boy. Finally, chromosome karyotype analysis confirmed that the boy had Klinefelter syndrome with a karyotype of 47, XXY. CONCLUSIONS: Klinefelter syndrome was accidentally detected during G6PD genetic analysis in a male. X-chromosomes can interfere with the results of G6PD genetic analysis and should be noted.


Sujet(s)
Déficit en glucose-6-phosphate-déshydrogénase , Glucose 6-phosphate dehydrogenase , Syndrome de Klinefelter , Humains , Syndrome de Klinefelter/génétique , Syndrome de Klinefelter/diagnostic , Syndrome de Klinefelter/complications , Mâle , Déficit en glucose-6-phosphate-déshydrogénase/génétique , Déficit en glucose-6-phosphate-déshydrogénase/complications , Déficit en glucose-6-phosphate-déshydrogénase/diagnostic , Glucose 6-phosphate dehydrogenase/génétique , Caryotypage , Mutation , Dépistage génétique/méthodes , Chromosomes X humains/génétique
11.
Sci Rep ; 14(1): 12802, 2024 06 04.
Article de Anglais | MEDLINE | ID: mdl-38834682

RÉSUMÉ

The presence of glucose-6-phosphate dehydrogenase (G6PD) deficiency may increase the risk of type 2 diabetes mellitus (T2DM), with differing prevalence between males and females. Although G6PD deficiency is an X-linked genetic condition, its interaction with sex regarding T2DM risk among the Taiwanese population has not been fully explored. This study aimed to investigate the association between G6PD deficiency and T2DM risk in the Taiwanese population, focusing on the potential influence of sex. Data were obtained from the Taiwan Biobank (TWB) database, involving 85,334 participants aged 30 to 70 years. We used multiple logistic regression analysis to assess the interaction between G6PD rs72554664 and sex in relation to T2DM risk. The T2DM cohort comprised 55.35% females and 44.65% males (p < 0.001). The TC + TT genotype of rs72554664 was associated with an increased risk of T2DM, with an odds ratio (OR) of 1.95 (95% CI: 1.39-2.75), and males showed an OR of 1.31 (95% CI: 1.19-1.44). Notably, the G6PD rs72554664-T allelic variant in hemizygous males significantly elevated the T2DM risk (OR), 4.57; p < 0.001) compared to females with the CC genotype. Our findings suggest that the G6PD rs72554664 variant, in conjunction with sex, significantly affects T2DM risk, particularly increasing susceptibility in males. The association of the G6PD rs72554664-T allelic variant with a higher risk of T2DM highlights the importance of sex-specific mechanisms in the interplay between G6PD deficiency and T2DM.


Sujet(s)
Biobanques , Diabète de type 2 , Prédisposition génétique à une maladie , Glucose 6-phosphate dehydrogenase , Polymorphisme de nucléotide simple , Humains , Mâle , Femelle , Adulte d'âge moyen , Taïwan/épidémiologie , Glucose 6-phosphate dehydrogenase/génétique , Diabète de type 2/génétique , Diabète de type 2/épidémiologie , Adulte , Sujet âgé , Déficit en glucose-6-phosphate-déshydrogénase/génétique , Déficit en glucose-6-phosphate-déshydrogénase/épidémiologie , Facteurs sexuels , Facteurs de risque , Génotype , Allèles
12.
J Biol Chem ; 300(7): 107460, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38876306

RÉSUMÉ

Obesity is a major risk factor for liver and cardiovascular diseases. However, obesity-driven mechanisms that contribute to the pathogenesis of multiple organ diseases are still obscure and treatment is inadequate. We hypothesized that increased , glucose-6-phosphate dehydrogenase (G6PD), the key rate-limiting enzyme in the pentose shunt, is critical in evoking metabolic reprogramming in multiple organs and is a significant contributor to the pathogenesis of liver and cardiovascular diseases. G6PD is induced by a carbohydrate-rich diet and insulin. Long-term (8 months) high-fat diet (HFD) feeding increased body weight and elicited metabolic reprogramming in visceral fat, liver, and aorta, of the wild-type rats. In addition, HFD increased inflammatory chemokines in visceral fat. Interestingly, CRISPR-edited loss-of-function Mediterranean G6PD variant (G6PDS188F) rats, which mimic human polymorphism, moderated HFD-induced weight gain and metabolic reprogramming in visceral fat, liver, and aorta. The G6PDS188F variant prevented HFD-induced CCL7 and adipocyte hypertrophy. Furthermore, the G6PDS188F variant increased Magel2 - a gene encoding circadian clock-related protein that suppresses obesity associated with Prader-Willi syndrome - and reduced HFD-induced non-alcoholic fatty liver. Additionally, the G6PDS188F variant reduced aging-induced aortic stiffening. Our findings suggest G6PD is a regulator of HFD-induced obesity, adipocyte hypertrophy, and fatty liver.


Sujet(s)
Adipocytes , Alimentation riche en graisse , Stéatose hépatique , Glucose 6-phosphate dehydrogenase , Hypertrophie , Obésité , Animaux , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/génétique , Mâle , Rats , Obésité/métabolisme , Obésité/génétique , Obésité/anatomopathologie , Obésité/étiologie , Alimentation riche en graisse/effets indésirables , Adipocytes/métabolisme , Adipocytes/anatomopathologie , Stéatose hépatique/métabolisme , Stéatose hépatique/génétique , Stéatose hépatique/anatomopathologie , Foie/métabolisme , Foie/anatomopathologie , Rat Sprague-Dawley , Graisse intra-abdominale/métabolisme , Graisse intra-abdominale/anatomopathologie
13.
Redox Biol ; 74: 103236, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38875958

RÉSUMÉ

The pathogenesis of epilepsy remains unclear; however, a prevailing hypothesis suggests that the primary underlying cause is an imbalance between neuronal excitability and inhibition. Glucose-6-phosphate dehydrogenase (G6PD) is a key enzyme in the pentose phosphate pathway, which is primarily involved in deoxynucleic acid synthesis and antioxidant defense mechanisms and exhibits increased expression during the chronic phase of epilepsy, predominantly colocalizing with neurons. G6PD overexpression significantly reduces the frequency and duration of spontaneous recurrent seizures. Furthermore, G6PD overexpression enhances signal transducer and activator of transcription 1 (STAT1) expression, thus influencing N-methyl-d-aspartic acid receptors expression, and subsequently affecting seizure activity. Importantly, the regulation of STAT1 by G6PD appears to be mediated primarily through reactive oxygen species signaling pathways. Collectively, our findings highlight the pivotal role of G6PD in modulating epileptogenesis, and suggest its potential as a therapeutic target for epilepsy.


Sujet(s)
Glucose 6-phosphate dehydrogenase , Espèces réactives de l'oxygène , Récepteurs du N-méthyl-D-aspartate , Facteur de transcription STAT-1 , Crises épileptiques , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/antagonistes et inhibiteurs , Glucose 6-phosphate dehydrogenase/génétique , Espèces réactives de l'oxygène/métabolisme , Animaux , Récepteurs du N-méthyl-D-aspartate/métabolisme , Récepteurs du N-méthyl-D-aspartate/génétique , Crises épileptiques/métabolisme , Crises épileptiques/traitement médicamenteux , Facteur de transcription STAT-1/métabolisme , Épilepsie/métabolisme , Épilepsie/traitement médicamenteux , Épilepsie/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Souris , Humains , Neurones/métabolisme , Mâle , Rats , Modèles animaux de maladie humaine
14.
Nat Commun ; 15(1): 5115, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38879607

RÉSUMÉ

Neurofibromatosis Type II (NFII) is a genetic condition caused by loss of the NF2 gene, resulting in activation of the YAP/TAZ pathway and recurrent Schwann cell tumors, as well as meningiomas and ependymomas. Unfortunately, few pharmacological options are available for NFII. Here, we undertake a genome-wide CRISPR/Cas9 screen to search for synthetic-lethal genes that, when inhibited, cause death of NF2 mutant Schwann cells but not NF2 wildtype cells. We identify ACSL3 and G6PD as two synthetic-lethal partners for NF2, both involved in lipid biogenesis and cellular redox. We find that NF2 mutant Schwann cells are more oxidized than control cells, in part due to reduced expression of genes involved in NADPH generation such as ME1. Since G6PD and ME1 redundantly generate cytosolic NADPH, lack of either one is compatible with cell viability, but not down-regulation of both. Since genetic deficiency for G6PD is tolerated in the human population, G6PD could be a good pharmacological target for NFII.


Sujet(s)
Systèmes CRISPR-Cas , Coenzyme A ligases , Glucose 6-phosphate dehydrogenase , Neurofibromine-2 , Cellules de Schwann , Mutations synthétiques létales , Cellules de Schwann/métabolisme , Humains , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/génétique , Neurofibromine-2/métabolisme , Neurofibromine-2/génétique , Coenzyme A ligases/métabolisme , Coenzyme A ligases/génétique , Animaux , Neurofibromatose de type 2/métabolisme , Neurofibromatose de type 2/génétique , NADP/métabolisme , Souris , Oxydoréduction
15.
Article de Anglais | MEDLINE | ID: mdl-38944269

RÉSUMÉ

The daily variations of temperature are one of the main synchronizers of the circadian rhythms. In addition, water temperature influences the embryonic and larval development of fish and directly affects their metabolic processes. The application of thermocycles to fish larvae has been reported to improve growth and the maturation of the digestive system, but their effects on metabolism are poorly understood. The aim of the present study was to evaluate the effect of two different temperature regimes, cycling versus constant, on the daily rhythms of metabolic factors of Nile tilapia (Oreochromis niloticus) larvae. For this purpose, fertilized eggs were divided into two groups: one reared in a 31 °C:25 °C day:night thermocycle (TCY) and another group maintained in a constant 28 °C temperature (CTE). The photoperiod was set to a 12:12 h light/dark cycle. Samples were collected every 4 h during a 24-h cycle on days 4, 8 and 13 post fertilization (dpf). The expression levels of alanine aminotransferase (alt), aspartate aminotransferase (ast), malic enzyme, glucose-6-phosphate dehydrogenase (g6pd), phosphofructokinase (pfk) and pyruvate kinase (pk) were analyzed by qPCR. Results showed that, in 13 dpf animals, most of the genes analyzed (alt, ast, malic, g6pd and pfk) showed daily rhythms in TCY, but not in the group kept at constant temperature, with most acrophases detected during the feeding period. An increase in nutrient metabolism around feeding time can improve food utilization and thus increase larval performance. Therefore, the use of thermocycles is recommended for tilapia larviculture.


Sujet(s)
Cichlides , Rythme circadien , Température , Animaux , Cichlides/croissance et développement , Cichlides/métabolisme , Cichlides/physiologie , Cichlides/génétique , Rythme circadien/physiologie , Larve/croissance et développement , Larve/métabolisme , Photopériode , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/génétique , Aspartate aminotransferases/métabolisme , Alanine transaminase/métabolisme
16.
FASEB J ; 38(10): e23705, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38805171

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive malignancies, with a notoriously dismal prognosis. As a competitive inhibitor of DNA synthesis, gemcitabine is the cornerstone drug for treating PDAC at all stages. The therapeutic effect of gemcitabine, however, is often hindered by drug resistance, and the underlying mechanisms remain largely unknown. It is unclear whether their response to chemotherapeutics is regulated by endocrine regulators, despite the association between PDAC risk and endocrine deregulation. Here, we show that prolactin receptor (PRLR) synergizes with gemcitabine in both in vitro and in vivo treatment of PDAC. Interestingly, PRLR promotes the expression of miR-4763-3p and miR-3663-5p, two novel miRNAs whose functions are unknown. Furthermore, the analysis of transcriptome sequencing data of tumors from lactating mouse models enriches the PPP pathway, a multifunctional metabolic pathway. In addition to providing energy, the PPP pathway mainly provides a variety of raw materials for anabolism. We demonstrate that two key enzymes of the pentose phosphate pathway (PPP), G6PD and TKT, are directly targeted by miR-4763-3p and miR-3663-5p. Notably, miR-4763-3p and miR-3663-5p diminish the nucleotide synthesis of the PPP pathway, thereby increasing gemcitabine sensitivity. As a result, PRLR harnesses these two miRNAs to suppress PPP and nucleotide synthesis, subsequently elevating the gemcitabine sensitivity of PDAC cells. Also, PDAC tissues and tumors from LSL-KrasG12D/+, LSL-Trp53R172H/+, and PDX1-cre (KPC) mice exhibit downregulation of PRLR. Bisulfite sequencing of PDAC tissues revealed that PRLR downregulation is due to epigenetic methylation. In this study, we show for the first time that the endocrine receptor PRLR improves the effects of gemcitabine by boosting two new miRNAs that block the PPP pathway and nucleotide synthesis by inhibiting two essential enzymes concurrently. The PRLR-miRNAs-PPP axis may serve as a possible therapeutic target to supplement chemotherapy advantages in PDAC.


Sujet(s)
Carcinome du canal pancréatique , Désoxycytidine , , Glucose 6-phosphate dehydrogenase , microARN , Tumeurs du pancréas , Récepteur prolactine , Animaux , Femelle , Humains , Souris , Antimétabolites antinéoplasiques/pharmacologie , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Lignée cellulaire tumorale , Désoxycytidine/analogues et dérivés , Désoxycytidine/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/génétique , microARN/génétique , microARN/métabolisme , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Récepteur prolactine/métabolisme , Récepteur prolactine/génétique , Souris nude
17.
Am J Trop Med Hyg ; 110(6): 1191-1197, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38593787

RÉSUMÉ

Glucose-6 phosphate dehydrogenase deficiency (G6PDd) was suggested as a risk factor for severe disease in patients with COVID-19. We evaluated clinical outcomes and glucose-6 phosphate dehydrogenase (G6PD) activity during and after illness in patients with COVID-19. This prospective cohort study included adult participants (≥ 18 years old) who had clinical and/or radiological COVID-19 findings or positive reverse transcription-polymerase chain reaction results. Epidemiological and clinical data were extracted from electronic medical records. Glucose-6 phosphate dehydrogenase activity was measured using SD Biosensor STANDARD G6PD® equipment on admission and 1 year after discharge. Samples were genotyped for the three most common single nucleotide polymorphisms for G6PDd in the Brazilian Amazon. Seven hundred fifty-three patients were included, of whom 123 (16.3%) were G6PD deficient. There was no difference between groups regarding the risks of hospitalization (P = 0.740) or invasive mechanical ventilation (P = 0.31), but the risk of death was greater in patients with normal G6PD levels (P = 0.022). Only 29 of 116 participants (25%) carried the African G6PDd genotype. Of 30 participants tested as G6PD deficient during disease, only 11 (36.7%) results agreed 1 year after discharge. In conclusion, this study does not demonstrate an association of G6PDd with severity of COVID-19. Limitations of the test for detecting enzyme levels during COVID-19 illness were demonstrated by genotyping and retesting after the disease period. Care must be taken when screening for G6PDd in patients with acute COVID-19.


Sujet(s)
COVID-19 , Déficit en glucose-6-phosphate-déshydrogénase , Glucose 6-phosphate dehydrogenase , SARS-CoV-2 , Adulte , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Brésil/épidémiologie , COVID-19/épidémiologie , Génotype , Glucose 6-phosphate dehydrogenase/génétique , Glucose 6-phosphate dehydrogenase/métabolisme , Déficit en glucose-6-phosphate-déshydrogénase/épidémiologie , Déficit en glucose-6-phosphate-déshydrogénase/génétique , Hospitalisation , Polymorphisme de nucléotide simple , Études prospectives , Facteurs de risque , SARS-CoV-2/génétique
18.
Sci China Life Sci ; 67(8): 1648-1665, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38679670

RÉSUMÉ

Betaine-homocysteine methyltransferase (BHMT) regulates protein methylation and is correlated with tumorigenesis; however, the effects and regulation of BHMT in hepatocarcinogenesis remain largely unexplored. Here, we determined the clinical significance of BHMT in the occurrence and progression of hepatocellular carcinoma (HCC) using tissue samples from 198 patients. BHMT was to be frequently found (86.6%) expressed at relatively low levels in HCC tissues and was positively correlated with the overall survival of patients with HCC. Bhmt overexpression effectively suppressed several malignant phenotypes in hepatoma cells in vitro and in vivo, whereas complete knockout of Bhmt (Bhmt-/-) produced the opposite effect. We combined proteomics, metabolomics, and molecular biological strategies and detected that Bhmt-/- promoted hepatocarcinogenesis and tumor progression by enhancing the activity of glucose-6-phosphate dehydrogenase (G6PD) and PPP metabolism in DEN-induced HCC mouse and subcutaneous tumor-bearing models. In contrast, restoration of Bhmt with an AAV8-Bhmt injection or pharmacological inhibition of G6PD attenuated hepatocarcinogenesis. Additionally, coimmunoprecipitation identified monomethylated modifications of the G6PD, and BHMT regulated the methylation of G6PD. Protein sequence analysis, generation and application of specific antibodies, and site-directed mutagenesis indicated G6PD methylation at the arginine residue 246. Furthermore, we established bidirectionally regulated BHMT cellular models combined with methylation-deficient G6PD mutants to demonstrate that BHMT potentiated arginine methylation of G6PD, thereby inhibiting G6PD activity, which in turn suppressed hepatocarcinogenesis. Taken together, this study reveals a new methylation-regulatory mechanism in hepatocarcinogenesis owing to BHMT deficiency, suggesting a potential therapeutic strategy for HCC treatment.


Sujet(s)
Betaine-homocysteine S-methyltransferase , Carcinogenèse , Carcinome hépatocellulaire , Glucose 6-phosphate dehydrogenase , Tumeurs du foie , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/métabolisme , Tumeurs du foie/génétique , Animaux , Humains , Méthylation , Betaine-homocysteine S-methyltransferase/métabolisme , Betaine-homocysteine S-methyltransferase/génétique , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/génétique , Souris , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Arginine/métabolisme , Mâle , Lignée cellulaire tumorale , Souris knockout , Régulation de l'expression des gènes tumoraux
19.
Sci Rep ; 14(1): 5806, 2024 03 09.
Article de Anglais | MEDLINE | ID: mdl-38461203

RÉSUMÉ

Due to the non-degradable and persistent nature of metal ions in the environment, they are released into water bodies, where they accumulate in fish. In order to assess pollution in fish, the enzyme, glucose 6-phosphate dehydrogenase (G6PD), has been employed as a biomarker due to sensitivity to various ions. This study investigates the kinetic properties of the G6PD enzyme in yellow catfish (Pelteobagrus fulvidraco), and analyzes the effects of these metal ions on the G6PD enzyme activity in the ovarian cell line (CCO) of channel catfish (Ictalurus punctatus). IC50 values and inhibition types of G6PD were determined in the metal ions Cu2+, Al3+, Zn2+, and Cd2+. While, the inhibition types of Cu2+ and Al3+ were the competitive inhibition, Zn2+ and Cd2+ were the linear mixed noncompetitive and linear mixed competitive, respectively. In vitro experiments revealed an inverse correlation between G6PD activity and metal ion concentration, mRNA levels and enzyme activity of G6PD increased at the lower metal ion concentration and decreased at the higher concentration. Our findings suggest that metal ions pose a significant threat to G6PD activity even at low concentrations, potentially playing a crucial role in the toxicity mechanism of metal ion pollution. This information contributes to the development of a biomonitoring tool for assessing metal ion contamination in aquatic species.


Sujet(s)
Cadmium , Poissons-chats , Animaux , Cadmium/toxicité , Cadmium/métabolisme , Métaux/pharmacologie , Métaux/métabolisme , Glucose 6-phosphate dehydrogenase/génétique , Poissons-chats/physiologie , Ions/métabolisme , Glucose/métabolisme , Phosphates/métabolisme
20.
Redox Biol ; 71: 103108, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38457903

RÉSUMÉ

High-risk human papillomaviruses (HPVs) are the causative agents of cervical cancer. Here, we report that HPV16 E6E7 promotes cervical cancer cell proliferation by activating the pentose phosphate pathway (PPP). We found that HPV16 E6 activates the PPP primarily by increasing glucose-6-phosphate dehydrogenase (G6PD) enzyme activity. Mechanistically, HPV16 E6 promoted G6PD dimer formation by inhibiting its lactylation. Importantly, we suggest that G6PD K45 was lactylated during G6PD-mediated antioxidant stress. In primary human keratinocytes and an HPV-negative cervical cancer C33A cells line ectopically expressing HPV16 E6, the transduction of G6PD K45A (unable to be lactylated) increased GSH and NADPH levels and, correspondingly, decreasing ROS levels. Conversely, the re-expression of G6PD K45T (mimicking constitutive lactylation) in HPV16-positive SiHa cells line inhibited cell proliferation. In vivo, the inhibition of G6PD enzyme activity with 6-aminonicotinamide (6-An) or the re-expression of G6PD K45T inhibited tumor proliferation. In conclusion, we have revealed a novel mechanism of HPV oncoprotein-mediated malignant transformation. These findings might provide effective strategies for treating cervical and HPV-associated cancers.


Sujet(s)
Protéines des oncogènes viraux , Infections à papillomavirus , Tumeurs du col de l'utérus , Femelle , Humains , Papillomavirus humain de type 16/génétique , Papillomavirus humain de type 16/métabolisme , Lignée cellulaire tumorale , Tumeurs du col de l'utérus/métabolisme , Glucose 6-phosphate dehydrogenase/génétique , Glucose 6-phosphate dehydrogenase/métabolisme , Voie des pentoses phosphates , Protéines des oncogènes viraux/génétique , Protéines des oncogènes viraux/métabolisme , Prolifération cellulaire
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE