RESUMO
BACKGROUND: During early pregnancy, glucose is essential for the uterine epithelium and the developing embryo. In cows, progesterone increases the secretion of glucose into the uterine lumen. The uterine epithelium can convert glucose to fructose, but other fates of glucose in the uterine epithelium have been sparsely investigated. Therefore, our objective was to investigate how progesterone influences glucose metabolism in immortalized bovine uterine epithelial (BUTE) cells. METHODS: BUTE cells were grown to 80% confluence and treated with vehicle (DMSO) or 10 µM progesterone for 24 h. Cells were collected and analyzed. Immunohistochemistry was performed on endometrial samples collected from the bovine endometrium on days 1 and 11 of the reproductive cycle. RESULTS: Progesterone treatment increased glucose consumption of BUTE cells. RNAseq identified 3,072 genes regulated by progesterone. KEGG analysis indicated that progesterone altered genes associated with metabolic pathways and glutathione metabolism. Manually examining genes unique to specific glucose metabolic pathways identified an increase in the rate-limiting enzyme in the pentose phosphate pathway-glucose-6-phosphate dehydrogenase. Functionally, a major product of the pentose phosphate pathway is NADPH, and progesterone treatment increased NADPH levels in BUTE cells. In cows, immunohistochemistry confirmed that glucose-6-phosphate dehydrogenase levels were higher in the uterine epithelium in the luteal phase when progesterone concentrations are high. CONCLUSIONS: Progesterone increased glucose-6-phosphate dehydrogenase expression and metabolism via the pentose phosphate pathway in the bovine uterine epithelium. This metabolism could provide substrates for cell proliferation, molecules to be secreted into the uterine lumen, or maintain reduction/oxidation balance in the uterine epithelium.
Assuntos
Endométrio , Células Epiteliais , Glucose , Glucosefosfato Desidrogenase , Via de Pentose Fosfato , Progesterona , Útero , Animais , Bovinos , Feminino , Via de Pentose Fosfato/efeitos dos fármacos , Progesterona/metabolismo , Progesterona/farmacologia , Células Epiteliais/metabolismo , Células Epiteliais/efeitos dos fármacos , Útero/metabolismo , Útero/efeitos dos fármacos , Glucosefosfato Desidrogenase/metabolismo , Glucosefosfato Desidrogenase/genética , Glucose/metabolismo , Endométrio/metabolismo , Endométrio/citologia , NADP/metabolismoRESUMO
The nanozyme with NADPH oxidase (NOX)-like activity can promote the consumption of NADPH and the generation of free radicals. In consideration of that the upregulation of glucose-6-phosphate dehydrogenase (G6PD) would accelerate the compensation production of NADPH, for inhibition of G6PD activity, our designed bioorthogonal nanozyme can in situ catalyze pro-DHEA to produce G6PD inhibitor and dehydroepiandrosterone (DHEA) drugs to inhibit G6PD activity. Therefore, the well-defined platform can disrupt NADPH homeostasis, leading to the collapse of the antioxidant defense system in the tumor cells. The enzyme-like activity of PdCuFe is further enhanced when irradiated by NIR-II light. The destruction of NADPH homeostasis can promote ferroptosis and, in turn, facilitate mild photothermal therapy. Our design can realize NADPH depletion and greatly improve the therapeutic effect through metabolic regulation, which may provide inspiration for the design of bioorthogonal catalysis.
Assuntos
Ferroptose , Glucosefosfato Desidrogenase , Terapia Fototérmica , Ferroptose/efeitos dos fármacos , Humanos , Glucosefosfato Desidrogenase/metabolismo , NADP/metabolismo , NADP/química , Animais , Camundongos , Desidroepiandrosterona/metabolismo , Desidroepiandrosterona/química , Desidroepiandrosterona/farmacologia , Antineoplásicos/farmacologia , Antineoplásicos/química , Nanopartículas/química , Nanopartículas/metabolismoRESUMO
Increased glycolytic metabolism recently emerged as an essential process driving host defense against Brucella, but little is known about how this process is regulated during infection. We have identified a critical role for nuclear factor kappa B (NF-κB) transcription factor regulation in glycolytic switching during Brucella infection for the first time. Chromatin immunoprecipitation with next-generation sequencing for NF-κB and DNA Pull-Down revealed two novel NF-κB-binding sites in the enhancer region of the Nitric oxide (NO)production-response regulator gene glucose-6-phosphate dehydrogenase (G6PD), which is important for the switch to glycolysis during a Brucella infection. These findings demonstrate that Brucella drives metabolic reprogramming by inhibiting host oxidative phosphorylation (OXPHOS) and enhancing its glycolysis via the NF-κB-G6PD-NO-pathway. These studies provide a theoretical basis for investigating drugs or vaccines to control Brucella colonization and induction of undulant by manipulating host metabolic patterns.
Assuntos
Brucelose , Glucosefosfato Desidrogenase , Glicólise , Macrófagos , NF-kappa B , Óxido Nítrico , Animais , Humanos , Camundongos , Brucella/imunologia , Brucelose/imunologia , Brucelose/metabolismo , Glucosefosfato Desidrogenase/metabolismo , Glucosefosfato Desidrogenase/genética , Células HEK293 , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Células RAW 264.7 , Transdução de Sinais , Regulação para CimaRESUMO
In the pentose phosphate pathway, dehydroepiandrosterone (DHEA) uncompetitively inhibits glucose-6-phosphate dehydrogenase (G6PD), reducing NADPH production and increasing oxidative stress, which can influence the onset and/or progression of several diseases, including cancer. 2-Deoxy-D-glucose (2-DG), a glucose mimetic, competes with glucose for cellular uptake, inhibiting glycolysis and competing with glucose-6-phosphate (G-6-P) for G6PD activity. In this study, we report that DHEA-α-2-DG (5), an α-covalent conjugate of DHEA and 2-DG, exhibits better anticancer activity than DHEA, 2-DG, DHEA +2-DG, and polydatin in MCF-7 cells, and reduces NADPH/NADP+ ratio in cellular assays. In vitro enzyme kinetics and molecular docking studies showed that 5 uncompetitively inhibits human G6PD activity and binds to the structural NADP+ site but not to the catalytic NADP+ site. Further combining 5 with the FDA-approved drug tamoxifen enhanced its cytotoxicity against MCF-7 cells, suggesting that it could serve as a candidate for combination of drug strategies.
Assuntos
Antineoplásicos , Neoplasias da Mama , Inibidores Enzimáticos , Glucosefosfato Desidrogenase , Feminino , Humanos , Antineoplásicos/farmacologia , Antineoplásicos/química , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Desidroepiandrosterona/farmacologia , Desidroepiandrosterona/química , Desoxiglucose/farmacologia , Desoxiglucose/química , Desoxiglucose/análogos & derivados , Desoxiglucose/metabolismo , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/química , Glucosefosfato Desidrogenase/metabolismo , Glucosefosfato Desidrogenase/antagonistas & inibidores , Cinética , Células MCF-7 , Simulação de Acoplamento Molecular , NADP/metabolismo , Tamoxifeno/farmacologia , Tamoxifeno/químicaRESUMO
BACKGROUND: G6PD (glucose-6-phosphate-dehydrogenase) is a key enzyme in the glycolytic pathway and has been implicated in the pathogenesis of cancer and pulmonary hypertension-associated vascular remodeling. Here, we investigated the role of an X-linked G6pd mutation (N126D polymorphism), which is known to increase the risk of cardiovascular disease in individuals from sub-Saharan Africa and many others with African ancestry, in the pathogenesis of pulmonary hypertension induced by a vascular endothelial cell growth factor receptor blocker used for treating cancer. METHODS AND RESULTS: CRISPR-Cas9 genome editing was used to generate the G6pd variant (N126D; G6pdN126D) in rats. A single dose of the vascular endothelial cell growth factor receptor blocker sugen-5416 (SU; 20 mg/kg in DMSO), which is currently in a Phase 2/3 clinical trial for cancer treatment, was subcutaneously injected into G6pdN126D rats and their wild-type littermates. After 8 weeks of normoxic conditions, right ventricular pressure and hypertrophy, pulmonary artery remodeling, the metabolic profile, and cytokine expression were assessed. Right ventricular pressure and pulmonary arterial wall thickness were increased in G6PDN126D+SU/normoxic rats. Simultaneously, levels of oxidized glutathione, inositol triphosphate, and intracellular Ca2+ were increased in the lungs of G6PDN126D+SU/normoxic rats, whereas nitric oxide was decreased. Also increased in G6PDN126D+SU/normoxic rats were pulmonary levels of plasminogen activator inhibitor-1, thrombin-antithrombin complex, and expression of proinflammatory cytokines CCL3 (chemokine [C-C motif] ligand), CCL5, and CCL7. CONCLUSIONS: Our results suggest G6PDN126D increases inositol triphosphate-Ca2+ signaling, inflammation, thrombosis, and hypertrophic pulmonary artery remodeling in SU-treated rats. This suggests an increased risk of vascular endothelial cell growth factor receptor blocker-induced pulmonary hypertension in those carrying this G6PD variant.
Assuntos
Glucosefosfato Desidrogenase , Receptores de Fatores de Crescimento do Endotélio Vascular , Animais , Glucosefosfato Desidrogenase/genética , Glucosefosfato Desidrogenase/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Ratos , Masculino , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Artéria Pulmonar/fisiopatologia , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/fisiopatologia , Modelos Animais de Doenças , Remodelação Vascular/efeitos dos fármacos , Ratos Sprague-Dawley , Indóis/farmacologia , Hipertrofia Ventricular Direita/genética , Hipertrofia Ventricular Direita/metabolismo , Hipertrofia Ventricular Direita/fisiopatologia , PirróisRESUMO
Glucose-6-phosphate dehydrogenase (G6PD) is the first enzyme in the pentose phosphate pathway. It has been extensively studied by biochemical and structural techniques. 13 X-ray crystal structures and five electron cryo-microscopy structures in the PDB are focused on in this topical review. Two F420-dependent glucose-6-phosphate dehydrogenase (FGD) structures are also reported. The significant differences between human and parasite G6PDs can be exploited to find selective drugs against infections such as malaria and leishmaniasis. Furthermore, G6PD is a prognostic marker in several cancer types and is also considered to be a tumour target. On the other hand, FGD is considered to be a target against Mycobacterium tuberculosis and possesses a high biotechnological potential in biocatalysis and bioremediation.
Assuntos
Microscopia Crioeletrônica , Glucosefosfato Desidrogenase , Modelos Moleculares , Microscopia Crioeletrônica/métodos , Cristalografia por Raios X , Humanos , Glucosefosfato Desidrogenase/química , Glucosefosfato Desidrogenase/metabolismo , Animais , Conformação Proteica , Mycobacterium tuberculosis/enzimologiaRESUMO
Cancer cells autonomously alter metabolic pathways in response to dynamic nutrient conditions in the microenvironment to maintain cell survival and proliferation. A better understanding of these adaptive alterations may reveal the vulnerabilities of cancer cells. Here, we demonstrate that coactivator-associated arginine methyltransferase 1 (CARM1) is frequently overexpressed in gastric cancer and predicts poor prognosis of patients with this cancer. Gastric cancer cells sense a reduced extracellular glucose content, leading to activation of nuclear factor erythroid 2-related factor 2 (NRF2). Subsequently, NRF2 mediates the classic antioxidant pathway to eliminate the accumulation of reactive oxygen species induced by low glucose. We found that NRF2 binds to the CARM1 promoter, upregulating its expression and triggering CARM1-mediated hypermethylation of histone H3 methylated at R arginine 17 (H3R17me2) in the glucose-6-phosphate dehydrogenase gene body. The upregulation of this dehydrogenase, driven by the H3R17me2 modification, redirects glucose carbon flux toward the pentose phosphate pathway. This redirection contributes to nucleotide synthesis (yielding nucleotide precursors, such as ribose-5-phosphate) and redox homeostasis and ultimately facilitates cancer cell survival and growth. NRF2 or CARM1 knockdown results in decreased H3R17me2a accompanied by the reduction of glucose-6-phosphate dehydrogenase under low glucose conditions. Collectively, this study reveals a significant role of CARM1 in regulating the tumor metabolic switch and identifies CARM1 as a potential therapeutic target for gastric cancer treatment.
Assuntos
Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Glucose , Fator 2 Relacionado a NF-E2 , Via de Pentose Fosfato , Proteína-Arginina N-Metiltransferases , Neoplasias Gástricas , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Humanos , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Via de Pentose Fosfato/genética , Glucose/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteína-Arginina N-Metiltransferases/genética , Linhagem Celular Tumoral , Animais , Glucosefosfato Desidrogenase/metabolismo , Glucosefosfato Desidrogenase/genética , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Histonas/metabolismo , Regiões Promotoras Genéticas/genética , Camundongos Nus , Transcrição Gênica , Proliferação de Células/genéticaRESUMO
BACKGROUND: Radical cure of Plasmodium vivax infections is key to the control of vivax malaria. However, the standard doses of 8-aminoquinoline drugs used for radical cure can cause severe haemolysis in G6PD-deficient patients. The availability of near-patient G6PD tests could increase use of primaquine (PQ), however direct evidence of the impacts that G6PD testing has on downstream patient outcomes, such as haemolysis and recurrence is lacking. METHODOLOGY/PRINCIPLE FINDINGS: A linked-evidence model was created to investigate changes in the number of severe haemolysis events and P. vivax recurrences within 6 months of treatment when qualitative G6PD testing was used to guide PQ treatment (0.25mg/kg/day for 14 days and 0.5mg/kg/day for 7 days), compared to prescribing 14-day PQ with no G6PD testing. In the model patients identified as G6PD-deficient received 8-week PQ (0.75mg/kg/week). The model was used to simulate scenarios with 1%, 5% and 10% prevalence of G6PD-deficiency (G6PDd) in theoretical populations of 10,000 male and female P. vivax patients and initially assumed 100% adherence to the prescribed PQ regiment. Results illustrate that G6PD testing to guide the 14-day PQ regiment reduced severe haemolysis by 21-80% and increased recurrences by 3-6%, compared to applying the 14-day PQ regiment without G6PD testing. Results for the 7-day PQ regiment informed by G6PD testing were mixed, dependent on G6PDd prevalence and sex. When adherence to the PQ regiments was less than perfect the model predicted reductions in the number of recurrences at all prevalence levels, provided adherence to 7-day PQ was 5-10% higher than adherence to the 14-day regiment. CONCLUSIONS/SIGNIFICANCE: Introduction of G6PD testing to guide PQ treatment reduces severe haemolysis events for the 14-day regiment, and the 7-day regiment in higher G6PDd prevalence settings, compared to use of 14-day PQ without G6PD testing when all patients adhere to the prescribed PQ treatment. At a population level, there were increases in recurrences, but this could be resolved when the 7-day regiment was used and had superior adherence compared to the 14-day regiment.
Assuntos
Antimaláricos , Deficiência de Glucosefosfato Desidrogenase , Hemólise , Malária Vivax , Plasmodium vivax , Primaquina , Humanos , Primaquina/uso terapêutico , Primaquina/administração & dosagem , Primaquina/efeitos adversos , Malária Vivax/tratamento farmacológico , Malária Vivax/parasitologia , Masculino , Feminino , Antimaláricos/uso terapêutico , Antimaláricos/administração & dosagem , Antimaláricos/efeitos adversos , Plasmodium vivax/efeitos dos fármacos , Deficiência de Glucosefosfato Desidrogenase/tratamento farmacológico , Adulto , Hemólise/efeitos dos fármacos , Adolescente , Adulto Jovem , Pessoa de Meia-Idade , Criança , Recidiva , Glucosefosfato Desidrogenase/metabolismo , Pré-EscolarRESUMO
Glioblastoma represents the predominant and a highly aggressive primary neoplasm of the central nervous system that has an abnormal metabolism. Our previous study showed that chrysomycin A (Chr-A) curbed glioblastoma progression in vitro and in vivo. However, whether Chr-A could inhibit orthotopic glioblastoma and how it reshapes metabolism are still unclear. In this study, Chr-A markedly suppressed the development of intracranial U87 gliomas. The results from airflow-assisted desorption electrospray ionization mass spectrometry imaging (AFADESI-MSI) indicated that Chr-A improved the abnormal metabolism of mice with glioblastoma. Key enzymes including glutaminase (GLS), glutamate dehydrogenases 1 (GDH1), hexokinase 2 (HK2) and glucose-6-phosphate dehydrogenase (G6PD) were regulated by Chr-A. Chr-A further altered the level of nicotinamide adenine dinucleotide phosphate (NADPH), thus causing oxidative stress with the downregulation of Nrf-2 to inhibit glioblastoma. Our study offers a novel perspective for comprehending the anti-glioma mechanism of Chr-A, highlighting its potential as a promising chemotherapeutic agent for glioblastoma.
Assuntos
Neoplasias Encefálicas , Glioblastoma , Estresse Oxidativo , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Humanos , Camundongos , Linhagem Celular Tumoral , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Glucosefosfato Desidrogenase/metabolismo , Antraquinonas/farmacologia , Glutaminase/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Progressão da Doença , Glutamato Desidrogenase/metabolismo , NADP/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Masculino , Camundongos NusRESUMO
Cold stress represents one of the major constraints for agricultural systems and crops productivity, inducing a wide range of negative effects. Particularly, long-term cold stress affects lipid metabolism, modifying the lipids/proteins ratio, the levels of phospholipids and glycolipids, and increasing lipids' unsaturation in bio-membranes. Glucose-6-phosphate dehydrogenase (G6PDH) reported prominent roles as NADPH suppliers in response to oxidative perturbations. Cytosolic G6PDH was suggested as the main isoform involved in cold stress response, while a down-regulation of the chloroplastic P1-G6PDH was observed. We thus investigated an Arabidopsis mutant defective for the P1-G6PDH (KO-P1) using integrated approaches to verify a possible role of this isoform in low temperature tolerance. KO-P1 genotype showed an improved tolerance to cold stress, highlighting a better photosynthetic efficiency, a reduction in stress markers content and a different regulation of genes involved in stress response. Intriguingly, the lack of P1-G6PDH induced modification in the levels of the main fatty acid and lipid species affecting the morphology of chloroplasts and mitochondria, which was restored under cold. Globally, these results indicate a priming effect induced by the absence of P1-G6PDH able to improve the tolerance to abiotic stress. Our results suggest novel and specific abilities of P1-G6PDH, highlighting its central role in different aspects of plant physiology and metabolism.
Assuntos
Arabidopsis , Cloroplastos , Resposta ao Choque Frio , Glucosefosfato Desidrogenase , Arabidopsis/genética , Arabidopsis/fisiologia , Arabidopsis/metabolismo , Glucosefosfato Desidrogenase/metabolismo , Glucosefosfato Desidrogenase/genética , Cloroplastos/metabolismo , Proteínas de Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Metabolismo dos Lipídeos , Regulação da Expressão Gênica de Plantas , Temperatura Baixa , Lipídeos/biossínteseRESUMO
Diabetic kidney disease (DKD) remains as one of the leading long-term complications of type 2 diabetic mellitus (T2DM). Studies have shown that decreased expression of glucose-6-phosphate dehydrogenase (G6PD) plays an important role in DKD. However, the upstream and downstream pathways of G6PD downregulation leading to DKD have not been elucidated. We conducted a series of studies including clinical study, animal studies, and in vitro studies to explore this. First, a total of 90 subjects were evaluated including 30 healthy subjects, 30 patients with T2DM, and 30 patients with DKD. The urinary G6PD activity and its association with the clinical markers were analyzed. Multivariate linear regression analysis was used to analyze the risk factors of urinary G6PD in these patients. Then, microRNAs that were differentially expressed in urine and could bind and degrade G6PD were screened and verified in patients with DKD. After that, high glucose (HG)-cultured human kidney cells (HK-2) and Zucker diabetic fatty (ZDF) rats were used to test the roles of miR-7977/G6PD/albumin-induced autophagy in DKD. Beclin and P62 were used as markers of kidney autophagy indicators. A dual-luciferase reporter assay system was used to test the binding of G6PD by mir-7977. The plasma and urinary G6PD activity were decreased significantly in patients with DKD, accompanied by increased urinary mir-7977 level. The fasting plasma glucose (FPG), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), and urinary albumin excretion were independent predictors of urinary G6PD activity, according to multiple linear regression analysis. The increased expression of miR-7977 and decreased expression of G6PD were also found in the kidney of ZDF rats with early renal tubular damage. The correlation analysis showed that beclin protein expression levels were positively correlated with kidney G6PD activity, whereas P62 protein expression was negatively correlated with kidney G6PD activity in rats. In HK-2 cells cultured with normal situation, a low level of albumin could induce autophagy along with the stimulation of G6PD, although this was impaired under high glucose. Overexpression of G6PD reversed albumin-induced autophagy in HK-2 cells under high glucose. Further study revealed that G6PD was a downstream target of miR-7977. Inhibition of miR-7977 expression led to significantly increased expression of G6PD and reversed the effects of high glucose on albumin-induced autophagy. In conclusion, our study supports a new mechanism of G6PD downregulation in DKD. Therapeutic measures targeting the miR-7977/G6PD/autophagy signaling pathway may help in the prevention and treatment of DKD.NEW & NOTEWORTHY This study provides new evidence that reduced glucose-6-phosphate dehydrogenase (G6PD) may damage the endocytosis of renal tubular epithelial cells by reducing albumin-induced autophagy. More importantly, for the first time, our study has provided evidence from humans that the decrease in urinary G6PD activity is positively associated with renal injury, and abnormal glucose and lipid metabolism may be important reasons for reduced G6PD levels. Increased miR-7977 may at least in part explain the downregulation of G6PD.
Assuntos
Autofagia , Diabetes Mellitus Tipo 2 , Nefropatias Diabéticas , Glucosefosfato Desidrogenase , MicroRNAs , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/urina , Autofagia/fisiologia , Masculino , Animais , Pessoa de Meia-Idade , Ratos , Glucosefosfato Desidrogenase/metabolismo , Glucosefosfato Desidrogenase/genética , Feminino , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/urina , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Ratos Zucker , Estudos de Casos e Controles , Células Epiteliais/metabolismo , Adulto , Linhagem Celular , IdosoRESUMO
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.
Assuntos
Glucosefosfato Desidrogenase , Morte Celular Imunogênica , Imunoterapia , Glucosefosfato Desidrogenase/metabolismo , Glucosefosfato Desidrogenase/antagonistas & inibidores , Animais , Humanos , Camundongos , Imunoterapia/métodos , Morte Celular Imunogênica/efeitos dos fármacos , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Feminino , Camundongos Endogâmicos C57BL , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Linhagem Celular Tumoral , Masculino , Melanoma/tratamento farmacológico , Melanoma/imunologia , Melanoma/patologiaRESUMO
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.
Assuntos
Progressão da Doença , Glucosefosfato Desidrogenase , Melaninas , Melanócitos , Sirtuínas , Vitiligo , Vitiligo/metabolismo , Vitiligo/patologia , Humanos , Melanócitos/metabolismo , Melanócitos/patologia , Sirtuínas/metabolismo , Sirtuínas/genética , Glucosefosfato Desidrogenase/metabolismo , Glucosefosfato Desidrogenase/genética , Melaninas/metabolismo , Melaninas/biossínteseRESUMO
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.
Assuntos
Óxido Nítrico , Nostoc , Nostoc/metabolismo , Nostoc/enzimologia , Nostoc/genética , Óxido Nítrico/metabolismo , Glucosefosfato Desidrogenase/metabolismo , Glucosefosfato Desidrogenase/genética , Polissacarídeos Bacterianos/metabolismo , Polissacarídeos Bacterianos/biossíntese , Polissacarídeos/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Escherichia coli/genética , Escherichia coli/metabolismoRESUMO
Glucose-6-phosphate dehydrogenase (G6PD) is a promising target in cancer therapy. However, poor cellular uptake and off-target toxicity have impeded the clinical translation of a canonical G6PD inhibitor (6-aminonicotinamide/6AN). Here, we report a prodrug strategy to address this issue. The tailored 6AN prodrug contains an azo-bearing protection moiety. The hydrophobic prodrug showed increased cellular uptake than 6AN and was vulnerable to hypoxia, resulting in NAD(P)H quinone dehydrogenase 1 (NQO1)-triggered cleavage of azo bonds. Intriguingly, the prodrug showed configuration-dependent anti-cancer potency. Despite the lower thermodynamic stability, the cis isomer showed enhanced cellular uptake compared to the trans counterpart due to the increased aqueous solubility. Moreover, the boosted potency of the cis isomer compared to the trans isomer arose from the enhancement of NOQ1-catalyzed 6AN release under hypoxia, a hallmark of solid tumors. The discovery of hypoxia-responsive 6AN prodrugs in the current work opens up new avenues for G6PD-targeting cancer medicines.
Assuntos
6-Aminonicotinamida , Antineoplásicos , NADP , Oxirredução , Pró-Fármacos , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Pró-Fármacos/síntese química , Humanos , 6-Aminonicotinamida/farmacologia , 6-Aminonicotinamida/química , NADP/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/síntese química , Proliferação de Células/efeitos dos fármacos , Estrutura Molecular , Glucosefosfato Desidrogenase/metabolismo , Glucosefosfato Desidrogenase/antagonistas & inibidores , Hipóxia Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos AntitumoraisRESUMO
Primaquine and Tafenoquine are the only approved drugs that can achieve a radical cure for Plasmodium vivax malaria but are contraindicated in patients who are deficient in glucose 6-phosphate dehydrogenase (G6PDd) due to risk of severe hemolysis from reactive oxygen species generated by redox cycling of drug metabolites. 5-hydroxyprimaquine and its quinoneimine cause robust redox cycling in red blood cells (RBCs) but are so labile as to not be detected in blood or urine. Rather, the quinoneimine is rapidly converted into primaquine-5,6-orthoquinone (5,6-POQ) that is then excreted in the urine. The extent to which 5,6-POQ contributes to hemolysis remains unclear, although some have suggested that it is a minor toxin that should be used predominantly as a surrogate to infer levels of 5-hydroxyprimaquine. In this report, we describe a novel humanized mouse model of the G6PD Mediterranean variant (hG6PDMed-) that recapitulates the human biology of RBC age-dependent enzyme decay, as well as an isogenic matched control mouse with human nondeficient G6PD hG6PDND In vitro challenge of RBCs with 5,6-POQ causes increased generation of superoxide and methemoglobin. Infusion of treated RBCs shows that 5,6-POQ selectively causes in vivo clearance of older hG6PDMed- RBCs. These findings support the hypothesis that 5,6-POQ directly induces hemolysis and challenges the notion that 5,6-POQ is an inactive metabolic waste product. Indeed, given the extreme lability of 5-hydroxyprimaquine and the relative stability of 5,6-POQ, these data raise the possibility that 5,6-POQ is a major hemolytic primaquine metabolite in vivo. SIGNIFICANCE STATEMENT: These findings demonstrate that 5,6-POQ, which has been considered an inert waste product of primaquine metabolism, directly induces ROS that cause clearance of older G6PDd RBCs. As 5,6-POQ is relatively stable compared with other active primaquine metabolites, these data support the hypothesis that 5,6-POQ is a major toxin in primaquine induced hemolysis. The findings herein also establish a new model of G6PDd and provide the first direct evidence, to our knowledge, that young G6PDd RBCs are resistant to primaquine-induced hemolysis.
Assuntos
Eritrócitos , Deficiência de Glucosefosfato Desidrogenase , Hemólise , Primaquina , Animais , Hemólise/efeitos dos fármacos , Eritrócitos/metabolismo , Eritrócitos/efeitos dos fármacos , Primaquina/farmacologia , Primaquina/metabolismo , Camundongos , Humanos , Deficiência de Glucosefosfato Desidrogenase/metabolismo , Glucosefosfato Desidrogenase/metabolismo , Modelos Animais de Doenças , Masculino , Antimaláricos/farmacologiaAssuntos
Complicações do Diabetes , Deficiência de Glucosefosfato Desidrogenase , Humanos , Deficiência de Glucosefosfato Desidrogenase/genética , Deficiência de Glucosefosfato Desidrogenase/complicações , Complicações do Diabetes/genética , Glucosefosfato Desidrogenase/genética , Glucosefosfato Desidrogenase/metabolismoRESUMO
The aim of the study was to investigate the relationship between ooplasm morphology, lipid content, glucose-6-phosphate dehydrogenase activity (G6PDH) and maturation potential of domestic cat oocytes. Cumulus-oocyte complexes were classified according to ooplasm morphology: evenly dark (dCOC), heterogeneous/mosaic (hCOC), or light/transparent (lCOC), however only dCOCs are thought to be the best-quality, the remaining ones are usually rejected, therefore little is known about their intracellular properties. Lipid droplets (LDs) were visualized and quantified using Oil Red O. G6PDH activity was assessed before in vitro maturation (IVM), using the brilliant cresyl blue (BCB) test. IVM-control oocytes underwent IVM without BCB staining. The dCOCs and hCOCs had different patterns of LD spatial distribution, but similar amounts of lipid, although this tended towards being lower in hCOCs. Low G6PDH activity (BCB+) was observed in 74 %, 60 % and 24 % (P < 0.01) of dCOCs, hCOCs, and lCOCs, respectively. Significantly more BCB+ /oocytes than BCB-/oocytes reached the metaphase II stage in all groups. The maturation rate of BCB+ /hCOCs was higher than that of IVM/hCOC-controls (40 % v.s. 20 %, P < 0.001), and was comparable to that of BCB+ /dCOCs (54 %; P > 0.05). lCOCs were the smallest (P < 0.01), contained fewer (P < 0.01) lipids than dCOCs or hCOCs, and displayed reduced maturational potential. Overall, LD content and distribution, as well as G6PDH activity, in cat oocytes were strongly associated with ooplasm morphology and oocyte maturational competence. Deeper understanding of the intrinsic properties of oocytes with different ooplasm morphology using the domestic cat model, may be particularly important in the context of the conservation of endangered felids.
Assuntos
Glucosefosfato Desidrogenase , Oócitos , Animais , Gatos , Oócitos/fisiologia , Glucosefosfato Desidrogenase/metabolismo , Feminino , Técnicas de Maturação in Vitro de Oócitos/veterinária , Células do Cúmulo/fisiologia , Células do Cúmulo/metabolismo , Metabolismo dos Lipídeos/fisiologia , Gotículas Lipídicas/metabolismo , LipídeosRESUMO
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.
Assuntos
COVID-19 , Deficiência de Glucosefosfato Desidrogenase , Lisossomos , Microglia , Óxido Nítrico Sintase Tipo II , Fagocitose , Humanos , COVID-19/metabolismo , COVID-19/patologia , COVID-19/virologia , COVID-19/imunologia , Glucosefosfato Desidrogenase/metabolismo , Glucosefosfato Desidrogenase/genética , Deficiência de Glucosefosfato Desidrogenase/metabolismo , Deficiência de Glucosefosfato Desidrogenase/patologia , Deficiência de Glucosefosfato Desidrogenase/genética , Lisossomos/metabolismo , Microglia/metabolismo , Microglia/patologia , NADP/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Células CultivadasRESUMO
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.