Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.718
Filtrer
1.
J Immunother Cancer ; 12(7)2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39089738

RÉSUMÉ

BACKGROUND: Enhanced glucose metabolism has been reported in many cancers. Glucose-6-phosphate dehydrogenase (G6PD) is a rate-limiting enzyme involved in the pentose phosphate pathway, which maintains NADPH levels and protects cells from oxidative damage. We recently found that low G6PD expression correlates with active tumor immunity. However, the mechanism involving G6PD and tumor immunity remained unclear. METHODS: We conducted in vitro studies using G6PD-knocked down malignant melanoma cells, pathway analysis using the GEO dataset, in vivo studies in combination with immune checkpoint inhibitors (ICIs) using a mouse melanoma model, and prognostic analysis in 42 melanoma patients and 30 lung cancer patients who were treated with ICIs. RESULTS: Inhibition of G6PD, both chemically and genetically, has been shown to decrease the production of NADPH and reduce their oxidative stress tolerance. This leads to cell death, which is accompanied by the release of high mobility group box 1 and the translocation of calreticulin to the plasma membrane. These findings suggested that inhibiting G6PD can induce immunogenic cell death. In experiments with C57BL/6 mice transplanted with G6PD-knockdown B16 melanoma cells and treated with anti-PD-L1 antibody, a significant reduction in tumor size was observed. Interestingly, inhibiting G6PD in only a part of the lesions increased the sensitivity of other lesions to ICI. Additionally, out of 42 melanoma patients and 30 lung cancer patients treated with ICIs, those with low G6PD expression had a better prognosis than those with high G6PD expression (p=0.0473; melanoma, p=0.0287; lung cancer). CONCLUSION: G6PD inhibition is a potent therapeutic strategy that triggers immunogenic cell death in tumors, significantly augmenting the efficacy of immunotherapies.


Sujet(s)
Glucose 6-phosphate dehydrogenase , Mort cellulaire immunogène , Immunothérapie , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/antagonistes et inhibiteurs , Animaux , Humains , Souris , Immunothérapie/méthodes , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Mélanome expérimental/traitement médicamenteux , Mélanome expérimental/immunologie , Mélanome expérimental/métabolisme , Femelle , Souris de lignée C57BL , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Lignée cellulaire tumorale , Mâle , Mélanome/traitement médicamenteux , Mélanome/immunologie , Mélanome/anatomopathologie
2.
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
3.
Ageing Res Rev ; 99: 102394, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38950868

RÉSUMÉ

Alzheimer's disease (AD) is a neurodegenerative pathologic entity characterized by the abnormal presence of tau and macromolecular Aß deposition that leads to the degeneration or death of neurons. In addition to that, glucose-6-phosphate dehydrogenase (G6PD) has a multifaceted role in the process of AD development, where it can be used as both a marker and a target. G6PD activity is dysregulated due to its contribution to oxidative stress, neuroinflammation, and neuronal death. In this context, the current review presents a vivid depiction of recent findings on the relationship between AD progression and changes in the expression or activity of G6PD. The efficacy of the proposed G6PD-based therapeutics has been demonstrated in multiple studies using AD mouse models as representative animal model systems for cognitive decline and neurodegeneration associated with this disease. Innovative therapeutic insights are made for the boosting of G6PD activity via novel innovative nanotechnology and microfluidics tools in drug administration technology. Such approaches provide innovative methods of surpassing the blood-brain barrier, targeting step-by-step specific neural pathways, and overcoming biochemical disturbances that accompany AD. Using different nanoparticles loaded with G6DP to target specific organs, e.g., G6DP-loaded liposomes, enhances BBB penetration and brain distribution of G6DP. Many nanoparticles, which are used for different purposes, are briefly discussed in the paper. Such methods to mimic BBB on organs on-chip offer precise disease modeling and drug testing using microfluidic chips, requiring lower sample amounts and producing faster findings compared to conventional techniques. There are other contributions to microfluid in AD that are discussed briefly. However, there are some limitations accompanying microfluidics that need to be worked on to be used for AD. This study aims to bridge the gap in understanding AD with the synergistic use of promising technologies; microfluid and nanotechnology for future advancements.


Sujet(s)
Maladie d'Alzheimer , Glucose 6-phosphate dehydrogenase , Nanoparticules , Maladie d'Alzheimer/métabolisme , Humains , Animaux , Glucose 6-phosphate dehydrogenase/métabolisme , Microfluidique/méthodes
4.
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
5.
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
6.
Biomed Pharmacother ; 176: 116935, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38876050

RÉSUMÉ

Breast cancer is one of the most common malignant tumors in women and is a serious threat to women's health. The pentose phosphate pathway (PPP) is a mode of oxidative breakdown of glucose that can be divided into oxidative (oxPPP) and non-oxidative (non-oxPPP) stages and is necessary for cell and body survival. However, abnormal activation of PPP often leads to proliferation, migration, invasion, and chemotherapy resistance in breast cancer. Glucose-6-phosphate dehydrogenase (G6PD) is the rate-limiting enzyme in PPP oxidation. Nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) produced by G6PD is the raw material for cholesterol and lipid synthesis and can resist the production of oxygen species (ROS) and reduce oxidative stress damage to tumor cells. Transketolase (TKT) is a key enzyme in non-oxPPP. Ribose 5-phosphate (R5P), produced by TKT, is a raw material for DNA and RNA synthesis, and is essential for tumor cell proliferation and DNA damage repair. In this review, we describe the role and specific mechanism of the PPP and the two most important enzymes of the PPP, G6PD and TKT, in the malignant progression of breast cancer, providing strategies for future clinical treatment of breast cancer and a theoretical basis for breast cancer research.


Sujet(s)
Tumeurs du sein , Évolution de la maladie , Glucose 6-phosphate dehydrogenase , Voie des pentoses phosphates , Transketolase , Transketolase/métabolisme , Humains , Tumeurs du sein/anatomopathologie , Tumeurs du sein/enzymologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Femelle , Glucose 6-phosphate dehydrogenase/métabolisme , Voie des pentoses phosphates/effets des médicaments et des substances chimiques , Animaux
7.
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
8.
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
9.
Front Immunol ; 15: 1393213, 2024.
Article de Anglais | MEDLINE | ID: mdl-38938571

RÉSUMÉ

Glucose-6-phosphate dehydrogenase (G6PD) deficiency is the most common enzymopathy in humans. G6PD is an essential enzyme in the pentose phosphate pathway (PPP), generating NADPH needed for cellular biosynthesis and reactive oxygen species (ROS) homeostasis, the latter especially key in red blood cells (RBCs). Beyond the RBC, there is emerging evidence that G6PD exerts an immunologic role by virtue of its functions in leukocyte oxidative metabolism and anabolic synthesis necessary for immune effector function. We review these here, and consider the global immunometabolic role of G6PD activity and G6PD deficiency in modulating inflammation and immunopathology.


Sujet(s)
Déficit en glucose-6-phosphate-déshydrogénase , Glucose 6-phosphate dehydrogenase , Humains , Glucose 6-phosphate dehydrogenase/métabolisme , Déficit en glucose-6-phosphate-déshydrogénase/immunologie , Déficit en glucose-6-phosphate-déshydrogénase/métabolisme , Animaux , Espèces réactives de l'oxygène/métabolisme , Voie des pentoses phosphates , Immunité , Infections/immunologie , Inflammation/immunologie , Inflammation/métabolisme
10.
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
11.
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
12.
Int J Biol Macromol ; 273(Pt 2): 132867, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38838892

RÉSUMÉ

Mounting an active immune response is energy intensive and demands the reallocation of nutrients to maintain the body's resistance and tolerance against infections. Central to this metabolic adaptation is Glucose-6-phosphate dehydrogenase (G6PDH), a housekeeping enzyme involve in pentose phosphate pathway (PPP). PPP play an essential role in generating ribose, which is critical for nicotinamide adenine dinucleotide phosphate (NADPH). It is vital for physiological and cellular processes such as generating nucleotides, fatty acids and reducing oxidative stress. The G6PDH is extremely conserved enzyme across species in PP shunt. The deficiency of enzymes leads to serious consequences on organism, particularly on adaptation and development. Acute deficiency can lead to impaired cell development, halted embryonic growth, reduce sensitivity to insulin, hypertension and increase inflammation. Historically, research focusing on G6PDH and PPP have primarily targeted diseases on mammalian. However, our review has investigated the unique functions of the G6PDH enzyme in insects and greatly improved mechanistic understanding of its operations. This review explore how G6PDH in insects plays a crucial role in managing the redox balance and immune related metabolism. This study aims to investigate the enzyme's role in different metabolic adaptations.


Sujet(s)
Glucose 6-phosphate dehydrogenase , Insectes , Oxydoréduction , Animaux , Glucose 6-phosphate dehydrogenase/métabolisme , Voie des pentoses phosphates , Stress oxydatif
13.
PLoS One ; 19(5): e0297918, 2024.
Article de Anglais | MEDLINE | ID: mdl-38728310

RÉSUMÉ

Quantitative diagnosis of glucose-6-phosphate dehydrogenase (G6PD) deficiency is essential for the safe administration of 8-aminoquinoline based radical cure for the treatment of Plasmodium vivax infections. Here, we present the PreQuine Platform (IVDS, USA), a quantitative biosensor that uses a dual-analyte assay for the simultaneous measurement of Hemoglobin (Hgb) levels and G6PD enzyme activity within the same sample. The platform relies on a downloadable mobile application. The device requires 10µl of whole blood and works with a reflectance-based meter. Comparing the G6PD measurement normalized by Hgb of 12 samples from the PreQuine Platform with reference measurements methods (spectrophotometry, Pointe Scientific, USA and hemoglobin meter, HemoCue, Sweden) showed a positive and significant agreement with a slope of 1.0091 and an intercept of -0.0379 under laboratory conditions. Next steps will be to conduct field trials in Bangladesh, Cambodia, and the USA to assess diagnostic performance, user friendliness and acceptance.


Sujet(s)
Déficit en glucose-6-phosphate-déshydrogénase , Glucose 6-phosphate dehydrogenase , Hémoglobines , Humains , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/sang , Déficit en glucose-6-phosphate-déshydrogénase/diagnostic , Déficit en glucose-6-phosphate-déshydrogénase/sang , Hémoglobines/analyse , Hémoglobines/métabolisme , Techniques de biocapteur/méthodes , Paludisme à Plasmodium vivax/diagnostic , Paludisme à Plasmodium vivax/sang , Aminoquinoléines
14.
Malar J ; 23(1): 140, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38725027

RÉSUMÉ

BACKGROUND: Plasmodium vivax relapses due to dormant liver hypnozoites can be prevented with primaquine. However, the dose must be adjusted in individuals with glucose-6-phosphate-dehydrogenase (G6PD) deficiency. In French Guiana, assessment of G6PD activity is typically delayed until day (D)14 to avoid the risk if misclassification. This study assessed the kinetics of G6PD activity throughout P. vivax infection to inform the timing of treatment. METHODS: For this retrospective monocentric study, data on G6PD activity between D1 and D28 after treatment initiation with chloroquine or artemisinin-based combination therapy were collected for patients followed at Cayenne Hospital, French Guiana, between January 2018 and December 2020. Patients were divided into three groups based on the number of available G6PD activity assessments: (i) at least two measurements during the P. vivax malaria infection; (ii) two measurements: one during the current infection and one previously; (iii) only one measurement during the malaria infection. RESULTS: In total, 210 patients were included (80, 20 and 110 in groups 1, 2 and 3, respectively). Data from group 1 showed that G6PD activity remained stable in each patient over time (D1, D3, D7, D14, D21, D28). None of the patients with normal G6PD activity during the initial phase (D1-D3) of the malaria episode (n = 44) was categorized as G6PD-deficient at D14. Patients with G6PD activity < 80% at D1 or D3 showed normal activity at D14. Sex and reticulocyte count were statistically associated with G6PD activity variation. In the whole sample (n = 210), no patient had severe G6PD deficiency (< 10%) and only three between 10 and 30%, giving a G6PD deficiency prevalence of 1.4%. Among the 100 patients from group 1 and 2, 30 patients (26.5%) were lost to follow-up before primaquine initiation. CONCLUSIONS: In patients treated for P. vivax infection, G6PD activity did not vary over time. Therefore, G6PD activity on D1 instead of D14 could be used for primaquine dose-adjustment. This could allow earlier radical treatment with primaquine, that could have a public health impact by decreasing early recurrences and patients lost to follow-up before primaquine initiation. This hypothesis needs to be confirmed in larger prospective studies.


Sujet(s)
Antipaludiques , Glucose 6-phosphate dehydrogenase , Paludisme à Plasmodium vivax , Adulte , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Jeune adulte , Antipaludiques/usage thérapeutique , Artémisinines/usage thérapeutique , Chloroquine/usage thérapeutique , Guyane française/épidémiologie , Glucose 6-phosphate dehydrogenase/métabolisme , Déficit en glucose-6-phosphate-déshydrogénase/épidémiologie , Déficit en glucose-6-phosphate-déshydrogénase/complications , Cinétique , Paludisme à Plasmodium vivax/traitement médicamenteux , Plasmodium vivax/effets des médicaments et des substances chimiques , Plasmodium vivax/physiologie , Primaquine/usage thérapeutique , Études rétrospectives , Sujet âgé de 80 ans ou plus
15.
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
16.
PLoS One ; 19(4): e0301506, 2024.
Article de Anglais | MEDLINE | ID: mdl-38687748

RÉSUMÉ

BACKGROUND: The WHO recommends routine testing of G6PD activity to guide radical cure in patients with Plasmodium vivax malaria. Females may have intermediate G6PD enzyme activity and to date, only complex diagnostics are able to reliably identify them. The semi-quantitative G6PD diagnostic "One Step G6PD Test" (Humasis, RoK; "RDT") is a lateral flow assay that can distinguish deficient, intermediate, and normal G6PD status and offers a simpler diagnostic alternative. METHODS: G6PD status of participants enrolled in Malinau and Nunukan Regencies and the capital Jakarta was assessed with the RDT, and G6PD activity was measured in duplicate by reference spectrophotometry. The adjusted male median (AMM) of the spectrophotometry measurements was defined as 100% activity; 70% and 30% of the AMM were defined as thresholds for intermediate and deficient G6PD status, respectively. Results were compared to those derived from spectrophotometry at the clinically relevant G6PD activity thresholds of 30% and 70%. RESULTS: Of the 161 participants enrolled, 10 (6.2%) were G6PD deficient and 12 (7.5%) had intermediate G6PD activity by spectrophotometry. At the 30% threshold, the sensitivity of the RDT was 10.0% (95%CI: 0.3-44.5%) with a specificity of 99.3% (95%CI: 96.4-100.0%); the positive predictive value was 50.0% (95%CI: 1.3-98.7%) and the negative predictive value 94.3% (95%CI: 89.5-97.4%). The corresponding figures at the 70% threshold were 22.7% (95%CI: 7.8-45.4%), 100.0% (95%CI: 97.4-100.0%), 100.0% (95%CI: 47.8-100.0%) and 89.1% (95%CI: 83.1-93.5%), respectively. CONCLUSION: While there is a dire need for an easy-to-use, economical, semi-quantitative diagnostic for the point of care, the observed performance of the "One Step G6PD Test" in its current form was insufficient to guide antimalarial treatment.


Sujet(s)
Déficit en glucose-6-phosphate-déshydrogénase , Paludisme à Plasmodium vivax , Humains , Déficit en glucose-6-phosphate-déshydrogénase/diagnostic , Femelle , Indonésie , Mâle , Adulte , Adolescent , Paludisme à Plasmodium vivax/diagnostic , Paludisme à Plasmodium vivax/sang , Adulte d'âge moyen , Jeune adulte , Systèmes automatisés lit malade , Enfant , Glucose 6-phosphate dehydrogenase/métabolisme , Glucose 6-phosphate dehydrogenase/sang , Spectrophotométrie/méthodes , Sensibilité et spécificité
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.
Cell Mol Biol Lett ; 29(1): 47, 2024 Apr 08.
Article de Anglais | MEDLINE | ID: mdl-38589823

RÉSUMÉ

BACKGROUND: Glucose-6-phosphate dehydrogenase (G6PD) plays an important role in vascular smooth muscle cell (VSMC) phenotypic switching, which is an early pathogenic event in various vascular remodeling diseases (VRDs). However, the underlying mechanism is not fully understood. METHODS: An IP‒LC‒MS/MS assay was conducted to identify new binding partners of G6PD involved in the regulation of VSMC phenotypic switching under platelet-derived growth factor-BB (PDGF-BB) stimulation. Co-IP, GST pull-down, and immunofluorescence colocalization were employed to clarify the interaction between G6PD and voltage-dependent anion-selective channel protein 1 (VDAC1). The molecular mechanisms involved were elucidated by examining the interaction between VDAC1 and apoptosis-related biomarkers, as well as the oligomerization state of VDAC1. RESULTS: The G6PD level was significantly elevated and positively correlated with the synthetic characteristics of VSMCs induced by PDGF-BB. We identified VDAC1 as a novel G6PD-interacting molecule essential for apoptosis. Specifically, the G6PD-NTD region was found to predominantly contribute to this interaction. G6PD promotes VSMC survival and accelerates vascular neointimal hyperplasia by inhibiting VSMC apoptosis. Mechanistically, G6PD interacts with VDAC1 upon stimulation with PDGF-BB. By competing with Bax for VDAC1 binding, G6PD reduces VDAC1 oligomerization and counteracts VDAC1-Bax-mediated apoptosis, thereby accelerating neointimal hyperplasia. CONCLUSION: Our study showed that the G6PD-VDAC1-Bax axis is a vital switch in VSMC apoptosis and is essential for VSMC phenotypic switching and neointimal hyperplasia, providing mechanistic insight into early VRDs.


Sujet(s)
Glucose 6-phosphate dehydrogenase , Muscles lisses vasculaires , Canal anionique-1 voltage-dépendant , Humains , Hyperplasie/métabolisme , Hyperplasie/anatomopathologie , Bécaplermine/génétique , Bécaplermine/métabolisme , Prolifération cellulaire , Protéine Bax/métabolisme , Glucose 6-phosphate dehydrogenase/métabolisme , Muscles lisses vasculaires/métabolisme , Chromatographie en phase liquide , Spectrométrie de masse en tandem , Néointima/génétique , Néointima/métabolisme , Néointima/anatomopathologie , Apoptose , Myocytes du muscle lisse/métabolisme , Mouvement cellulaire/génétique , Cellules cultivées , Phénotype
19.
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
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