Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 417
Filtrar
1.
Front Biosci (Landmark Ed) ; 29(1): 27, 2024 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-38287804

RESUMO

BACKGROUND: The pentose phosphate pathway (PPP) is a critical metabolic pathway that generates NADPH and ribose-5-phosphate for nucleotide biosynthesis and redox homeostasis. In this study, we investigated a potential regulatory role for Krüppel-like factor 8 (KLF8) in the control of PPP in lung adenocarcinoma (LUAD) cells. METHODS: Based on a comprehensive set of experimental approaches, including cell culture, molecular techniques, and functional assays, we revealed a novel mechanism by which KLF8 promotes the activation of glucose-6-phosphate dehydrogenase (G6PD), a component enzyme in the PPP. RESULTS: Our findings demonstrate that KLF8 inhibits the acetylation of G6PD, leading to its increased enzymatic activity. Additionally, we observed that KLF8 activates the transcription of SIRT2, which has been implicated in regulating G6PD acetylation. These results highlight the interplay between KLF8, G6PD, and protein acetylation in the regulation of PPP in LUAD. CONCLUSIONS: Understanding the intricate molecular mechanisms underlying the metabolic reprogramming driven by KLF8 in lung cancer provides valuable insights into potential therapeutic strategies targeting the PPP. This study emphasizes the significance of KLF8 as a key modulator of metabolic pathways and indicates the potential of targeting the KLF8-G6PD axis for lung cancer treatment.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Humanos , Via de Pentose Fosfato/fisiologia , Sirtuína 2/genética , Sirtuína 2/metabolismo , Adenocarcinoma de Pulmão/genética , Neoplasias Pulmonares/genética , Pulmão/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo
2.
Chem Res Toxicol ; 37(2): 208-211, 2024 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-38191130

RESUMO

The Cell Counting Kit-8 (CCK-8) cell viability assay, also known as WST-8, is widely recognized for its nontoxic nature, making it suitable for further studies on treated cells. This practice is commonly observed in the field of tissue engineering. While live/dead imaging may not readily reveal macroscopic differences, our investigation has uncovered significant intracellular metabolic changes. Notably, we observed substantial down-regulation of metabolites within the glycolysis and pentose phosphate pathways. These metabolic alterations predominantly affect energy metabolism and may potentially impact the cellular redox environment. In light of these findings, we strongly recommend that researchers exercise caution when using cells treated with CCK-8 in subsequent experiments.


Assuntos
Glicólise , Via de Pentose Fosfato , Via de Pentose Fosfato/fisiologia , Sobrevivência Celular , Glicólise/fisiologia , Metabolismo Energético , Metaboloma
3.
Biochem Soc Trans ; 51(6): 2173-2187, 2023 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-37971161

RESUMO

The pentose phosphate pathway (PPP) is a key metabolic pathway. The oxidative phase of this process involves three reactions catalyzed by glucose-6-phosphate dehydrogenase (G6PDH), 6-phosphogluconolactonase (6PGL) and 6-phosphogluconate dehydrogenase (6PGDH) enzymes. The first and third steps (catalyzed by G6PDH and 6PGDH, respectively) are responsible for generating reduced nicotinamide adenine dinucleotide phosphate (NAPDH), a key cofactor for maintaining the reducing power of cells and detoxification of both endogenous and exogenous oxidants and electrophiles. Despite the importance of these enzymes, little attention has been paid to the fact that these proteins are targets of oxidants. In response to oxidative stimuli metabolic pathways are modulated, with the PPP often up-regulated in order to enhance or maintain the reductive capacity of cells. Under such circumstances, oxidation and inactivation of the PPP enzymes could be detrimental. Damage to the PPP enzymes may result in a downward spiral, as depending on the extent and sites of modification, these alterations may result in a loss of enzymatic activity and therefore increased oxidative damage due to NADPH depletion. In recent years, it has become evident that the three enzymes of the oxidative phase of the PPP have different susceptibilities to inactivation on exposure to different oxidants. In this review, we discuss existing knowledge on the role that these enzymes play in the metabolism of cells, and their susceptibility to oxidation and inactivation with special emphasis on NADPH production. Perspectives on achieving a better understanding of the molecular basis of the oxidation these enzymes within cellular environments are given.


Assuntos
Estresse Oxidativo , Via de Pentose Fosfato , Via de Pentose Fosfato/fisiologia , NADP/química , NADP/metabolismo , Oxirredução , Oxidantes
4.
Biochem Pharmacol ; 217: 115856, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37838274

RESUMO

Maintaining redox homeostasis is an essential feature of cancer cells, and disrupting this homeostasis to cause oxidative stress and induce cell death is an important strategy in cancer therapy. M4IDP, a zoledronic acid derivative, can cause the death of human colorectal cancer cells by increasing the level of intracellular reactive oxygen species (ROS). However, its potential molecular mechanism is unclear. Our in vitro studies showed that treatment with M4IDP promoted oxidative stress in HCT116 cells, as measured by the decreased ratios of GSH/GSSG and NADPH/NADP+ and increased level of MDA. M4IDP could cause the decrease of GSH content, the increase of GSSG content, the decrease of NADPH content and pentose phosphate pathway flux, the downregulation of G6PD expression, the upregulation of unprenylated Rap1A and total expression of RhoA and CDC42. The increase of ROS and cytotoxicity induced by M4IDP could be reversed by the supplementation of NADPH, the overexpression of G6PD and the supplementation of GGOH. In vivo studies showed that M4IDP inhibited tumor growth in the human colorectal cancer xenograft mouse model, which was accompanied with a decreased [18F]FDG uptake. Collectively, these results provide evidence that M4IDP can promote oxidation in colon cancer cells by inhibiting mevalonate pathway and pentose phosphate pathway and produce therapeutic effect. This study revealed for the first time a possible mechanism of bisphosphonate-induced increase of ROS in malignant tumor cells. This is helpful for the development of new molecular therapeutic targets and can provide new ideas for the combined therapy of bisphosphonates in tumors.


Assuntos
Neoplasias do Colo , Ácido Mevalônico , Humanos , Camundongos , Animais , Espécies Reativas de Oxigênio/metabolismo , Dissulfeto de Glutationa/metabolismo , Dissulfeto de Glutationa/farmacologia , Ácido Mevalônico/farmacologia , NADP/metabolismo , NADP/farmacologia , Via de Pentose Fosfato/fisiologia , Estresse Oxidativo , Neoplasias do Colo/tratamento farmacológico
5.
Neurochem Res ; 48(10): 3177-3189, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37394677

RESUMO

Glucose-6-phosphate dehydrogenase (G6PDH) catalyses the rate limiting first step of the oxidative part of the pentose phosphate pathway (PPP), which has a crucial function in providing NADPH for antioxidative defence and reductive biosyntheses. To explore the potential of the new G6PDH inhibitor G6PDi-1 to affect astrocytic metabolism, we investigated the consequences of an application of G6PDi-1 to cultured primary rat astrocytes. G6PDi-1 efficiently inhibited G6PDH activity in lysates of astrocyte cultures. Half-maximal inhibition was observed for 100 nM G6PDi-1, while presence of almost 10 µM of the frequently used G6PDH inhibitor dehydroepiandrosterone was needed to inhibit G6PDH in cell lysates by 50%. Application of G6PDi-1 in concentrations of up to 100 µM to astrocytes in culture for up to 6 h did not affect cell viability nor cellular glucose consumption, lactate production, basal glutathione (GSH) export or the high basal cellular ratio of GSH to glutathione disulfide (GSSG). In contrast, G6PDi-1 drastically affected astrocytic pathways that depend on the PPP-mediated supply of NADPH, such as the NAD(P)H quinone oxidoreductase (NQO1)-mediated WST1 reduction and the glutathione reductase-mediated regeneration of GSH from GSSG. These metabolic pathways were lowered by G6PDi-1 in a concentration-dependent manner in viable astrocytes with half-maximal effects observed for concentrations between 3 and 6 µM. The data presented demonstrate that G6PDi-1 efficiently inhibits the activity of astrocytic G6PDH and impairs specifically those metabolic processes that depend on the PPP-mediated regeneration of NADPH in cultured astrocytes.


Assuntos
Astrócitos , Via de Pentose Fosfato , Ratos , Animais , Astrócitos/metabolismo , Via de Pentose Fosfato/fisiologia , Dissulfeto de Glutationa/metabolismo , Glucosefosfato Desidrogenase/metabolismo , NADP/metabolismo , Glutationa/metabolismo , Células Cultivadas
6.
Proc Natl Acad Sci U S A ; 120(4): e2212813120, 2023 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-36649417

RESUMO

The immune system must be able to respond to a myriad of different threats, each requiring a distinct type of response. Here, we demonstrate that the cytoplasmic lysine deacetylase HDAC7 in macrophages is a metabolic switch that triages danger signals to enable the most appropriate immune response. Lipopolysaccharide (LPS) and soluble signals indicating distal or far-away danger trigger HDAC7-dependent glycolysis and proinflammatory IL-1ß production. In contrast, HDAC7 initiates the pentose phosphate pathway (PPP) for NADPH and reactive oxygen species (ROS) production in response to the more proximal threat of nearby bacteria, as exemplified by studies on uropathogenic Escherichia coli (UPEC). HDAC7-mediated PPP engagement via 6-phosphogluconate dehydrogenase (6PGD) generates NADPH for antimicrobial ROS production, as well as D-ribulose-5-phosphate (RL5P) that both synergizes with ROS for UPEC killing and suppresses selective inflammatory responses. This dual functionality of the HDAC7-6PGD-RL5P axis prioritizes responses to proximal threats. Our findings thus reveal that the PPP metabolite RL5P has both antimicrobial and immunomodulatory activities and that engagement of enzymes in catabolic versus anabolic metabolic pathways triages responses to different types of danger for generation of inflammatory versus antimicrobial responses, respectively.


Assuntos
Anti-Infecciosos , Triagem , Espécies Reativas de Oxigênio/metabolismo , NADP/metabolismo , Macrófagos/metabolismo , Anti-Infecciosos/metabolismo , Via de Pentose Fosfato/fisiologia
7.
FEBS J ; 290(3): 763-779, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36048131

RESUMO

Dysregulation of G6PD involved in the pentose phosphate pathway (PPP) is known to promote tumorigenesis. The PPP plays a pivotal role in meeting the anabolic demands of cancer cells. However, the detailed underlying molecular mechanisms of targeting the G6PD-regulated PPP in breast cancer remain unclear. In this study, we aimed to elucidate the molecular pathways mediating the effects of G6PD on cancer progression. Clinical sample analysis found that the expression of G6PD in breast cancer patients was higher than that in normal controls, and patients with higher G6PD expression had poor survival. Gene knockdown or inhibition of G6PD by 6-AN in MCF-7 and MDA-MB-231 cells significantly decreased cell viability, migration, and colony formation ability. G6PD enzyme activity was inhibited by 6-AN treatment, which caused a transient upregulation of ROS. The elevated ROS was independent of cell apoptosis and thus associated with abnormal activated autophagy. Accumulated ROS levels induced autophagic cell death in breast cancer. Inhibition of G6PD suppresses tumour growth in preclinical models of breast cancer. Our results indicate that targeting the G6PD-regulated PPP could restrain tumours in vitro and in vivo, inhibiting G6PD caused cell death by over-activating autophagy, therefore leading to inhibited proliferation and tumour formation.


Assuntos
Morte Celular Autofágica , Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/patologia , Espécies Reativas de Oxigênio/metabolismo , Glucosefosfato Desidrogenase/genética , Apoptose , Via de Pentose Fosfato/fisiologia , Linhagem Celular Tumoral , Proliferação de Células
8.
PLoS Pathog ; 18(9): e1010864, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36121870

RESUMO

Metabolic pathways underpin the growth and virulence of intracellular parasites and are therefore promising antiparasitic targets. The pentose phosphate pathway (PPP) is vital in most organisms, providing a reduced form of nicotinamide adenine dinucleotide phosphate (NADPH) and ribose sugar for nucleotide synthesis; however, it has not yet been studied in Toxoplasma gondii, a widespread intracellular pathogen and a model protozoan organism. Herein, we show that T. gondii has a functional PPP distributed in the cytoplasm and nucleus of its acutely-infectious tachyzoite stage. We produced eight parasite mutants disrupting seven enzymes of the PPP in T. gondii. Our data show that of the seven PPP proteins, the two glucose-6-phosphate dehydrogenases (TgG6PDH1, TgG6PDH2), one of the two 6-phosphogluconate dehydrogenases (Tg6PGDH1), ribulose-5-phosphate epimerase (TgRuPE) and transaldolase (TgTAL) are dispensable in vitro as well as in vivo, disclosing substantial metabolic plasticity in T. gondii. Among these, TgG6PDH2 plays a vital role in defense against oxidative stress by the pathogen. Further, we show that Tg6PGDH2 and ribulose-5-phosphate isomerase (TgRPI) are critical for tachyzoite growth. The depletion of TgRPI impairs the flux of glucose in central carbon pathways, and causes decreased expression of ribosomal, microneme and rhoptry proteins. In summary, our results demonstrate the physiological need of the PPP in T. gondii while unraveling metabolic flexibility and antiparasitic targets.


Assuntos
Via de Pentose Fosfato , Toxoplasma , Antiparasitários , Carbono/metabolismo , Glucose/metabolismo , Glucose-6-Fosfato/metabolismo , Isomerases/metabolismo , NADP/metabolismo , Via de Pentose Fosfato/fisiologia , Fosfatos/metabolismo , Racemases e Epimerases/metabolismo , Ribose , Toxoplasma/metabolismo , Transaldolase/metabolismo
9.
Biol Reprod ; 107(4): 998-1013, 2022 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-35717588

RESUMO

Establishing an ideal human follicle culture system for oncofertility patients relies mainly on animal models since donor tissue is scarce and often of suboptimal quality. The in vitro system developed in our laboratory supports the growth of prepubertal mouse secondary follicles up to mature oocytes. Given the importance of glucose in preparing the oocyte for proper maturation, a baseline characterization of follicle metabolism both in the culture system and in vivo was carried out. Markers of glucose-related pathways (glycolysis, tricarboxylic acid [TCA] cycle, pentose phosphate pathway [PPP], polyol pathway, and hexosamine biosynthetic pathway), as well as the antioxidant capacity, were measured in the different follicle cell types by both enzymatic activities (spectrophotometric detection) and gene expression (qPCR). This study confirmed that in vivo the somatic cells, mainly granulosa, exhibit intense glycolytic activity, while oocytes perform PPP. Throughout the final maturation step, oocytes in vivo and in vitro showed steady levels for all the key enzymes and metabolites. On the other hand, ovulation triggers a boost of pyruvate and lactate uptake in cumulus cells in vivo, consumes reduced nicotinamide adenine dinucleotide phosphate, and increases TCA cycle and small molecules antioxidant capacity activities, while in vitro, the metabolic upregulation in all the studied pathways is limited. This altered metabolic pattern might be a consequence of cell exhaustion because of culture conditions, impeding cumulus cells to fulfill their role in providing proper support for acquiring oocyte competence.


Assuntos
Antioxidantes , Oócitos , Animais , Antioxidantes/metabolismo , Células do Cúmulo/metabolismo , Feminino , Glucose/metabolismo , Hexosaminas/metabolismo , Humanos , Ácido Láctico/metabolismo , Camundongos , NADP/metabolismo , Oócitos/metabolismo , Via de Pentose Fosfato/fisiologia , Ácido Pirúvico/metabolismo , Ácidos Tricarboxílicos/metabolismo
10.
Brain Res Bull ; 185: 162-173, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35588962

RESUMO

Subarachnoid hemorrhage (SAH), a type of hemorrhagic stroke, is a neurological emergency with high morbidity and mortality. Early brain injury (EBI) after SAH is the leading cause of poor prognosis in SAH patients. TRX system is a NADPH-dependent antioxidant system which is composed of thioredoxin reductase (TRXR), thioredoxin (TRX). The pentose phosphate pathway (PPP), a pathway through which glucose can be metabolized, is a major source of NADPH. Thioredoxin 1 (TRX1) is a member of thioredoxin system mainly located in cytoplasm. Serine/threonine kinases ataxia telangiectasia mutated (ATM) is an important oxidative stress receptor, and TRX1 can regulate ATM phosphorylation and then affect the activity of PPP key enzyme glucose 6-phosphate dehydrogenase (G6PD). However, whether TRX1 is involved in the regulation of PPP pathway after subarachnoid hemorrhage remains unclear. The results showed that after SAH, the level of TRX1 and phosphor-ATM decreased while the level of TRXR1 increased. G6PD protein level remained unchanged but the activity decreased, and the NADPH contents decreased. Overexpression of TRX1 by lentivirus upregulates the level of phosphor-ATM, G6PD activity and NADPH content. TRX1 overexpression improved short-term and long-term neurobehavioral outcomes and alleviated neuronal impairment in rats. Nissl staining showed that upregulation of TRX1 reduced cortical neuron injury. Our study shows that TRX1 participates in the PPP pathway by regulating phosphorylation ATM, which is accomplished by affecting G6PD activity. TRX1 may be an important target for EBI intervention after SAH.


Assuntos
Ataxia Telangiectasia , Hemorragia Subaracnóidea , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Glucose , Humanos , NADP/metabolismo , Via de Pentose Fosfato/fisiologia , Fosforilação , Ratos , Tiorredoxinas/metabolismo
11.
Sci Rep ; 12(1): 4601, 2022 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-35301361

RESUMO

Due to its role in maintaining the proper functioning of the cell, the pentose phosphate pathway (PPP) is one of the most important metabolic pathways. It is responsible for regulating the concentration of simple sugars and provides precursors for the synthesis of amino acids and nucleotides. In addition, it plays a critical role in maintaining an adequate level of NADPH, which is necessary for the cell to fight oxidative stress. These reasons prompted the authors to develop a computational model, based on queueing theory, capable of simulating changes in PPP metabolites' concentrations. The model has been validated with empirical data from tumor cells. The obtained results prove the stability and accuracy of the model. By applying queueing theory, this model can be further expanded to include successive metabolic pathways. The use of the model may accelerate research on new drugs, reduce drug costs, and reduce the reliance on laboratory animals necessary for this type of research on which new methods are tested.


Assuntos
Estresse Oxidativo , Via de Pentose Fosfato , Animais , NADP/metabolismo , Via de Pentose Fosfato/fisiologia
12.
Expert Opin Drug Metab Toxicol ; 18(1): 61-83, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35238253

RESUMO

INTRODUCTION: The pentose phosphate pathway (PPP) branches from glycolysis and is crucial for cell growth, since it provides necessary compounds for anabolic reactions, nucleotide synthesis, and detoxification of reactive-oxygen-species (ROS). Overexpression of PPP enzymes has been reported in multiple cancer types and linked to therapy resistance, making their inhibition interesting targets for anti-cancer therapies. AREAS COVERED: This review summarizes the extent of PPP upregulation across different cancer types, and the non-metabolic functions that PPP-enzymes might contribute to cancer initiation and maintenance. The effects of PPP-inhibition and their combinations with chemotherapeutics are summarized. We searched the databases provided by the University of Amsterdam to characterize the altered expression of the PPP across different cancer types, and to identify the effects of PPP-inhibition. EXPERT OPINION: It can be concluded that there are synergistic and additive effects of PPP-inhibition and various classes of chemotherapeutics. These effects may be attributed to the increased susceptibility to ROS. However, the toxicity, low efficacy, and off-target effects of PPP-inhibitors make application in clinical practice challenging. Novel inhibitors are currently being developed, which could make PPP-inhibition a potential therapeutic strategy in the future, especially in combination with conventional chemotherapeutics and the inhibition of other metabolic pathways.


Assuntos
Neoplasias , Via de Pentose Fosfato , Proliferação de Células , Glicólise/fisiologia , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Via de Pentose Fosfato/fisiologia , Espécies Reativas de Oxigênio/metabolismo
13.
Sci Adv ; 8(8): eabf9096, 2022 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-35213227

RESUMO

The spread of cancer to bone is invariably fatal, with complex cross-talk between tumor cells and the bone microenvironment responsible for driving disease progression. By combining in silico analysis of patient datasets with metabolomic profiling of prostate cancer cells cultured with bone cells, we demonstrate the changing energy requirements of prostate cancer cells in the bone microenvironment, identifying the pentose phosphate pathway (PPP) as elevated in prostate cancer bone metastasis, with increased expression of the PPP rate-limiting enzyme glucose-6-phosphate dehydrogenase (G6PD) associated with a reduction in progression-free survival. Genetic and pharmacologic manipulation demonstrates that G6PD inhibition reduces prostate cancer growth and migration, associated with changes in cellular redox state and increased chemosensitivity. Genetic blockade of G6PD in vivo results in reduction of tumor growth within bone. In summary, we demonstrate the metabolic plasticity of prostate cancer cells in the bone microenvironment, identifying the PPP and G6PD as metabolic targets for the treatment of prostate cancer bone metastasis.


Assuntos
Glucosefosfato Desidrogenase , Neoplasias da Próstata , Linhagem Celular Tumoral , Glucosefosfato Desidrogenase/genética , Glucosefosfato Desidrogenase/metabolismo , Humanos , Masculino , Metabolômica , Via de Pentose Fosfato/fisiologia , Neoplasias da Próstata/genética , Microambiente Tumoral
14.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35110412

RESUMO

The pentose phosphate pathway is a major source of NADPH for oxidative stress resistance in cancer cells but there is limited insight into its role in metastasis, when some cancer cells experience high levels of oxidative stress. To address this, we mutated the substrate binding site of glucose 6-phosphate dehydrogenase (G6PD), which catalyzes the first step of the pentose phosphate pathway, in patient-derived melanomas. G6PD mutant melanomas had significantly decreased G6PD enzymatic activity and depletion of intermediates in the oxidative pentose phosphate pathway. Reduced G6PD function had little effect on the formation of primary subcutaneous tumors, but when these tumors spontaneously metastasized, the frequency of circulating melanoma cells in the blood and metastatic disease burden were significantly reduced. G6PD mutant melanomas exhibited increased levels of reactive oxygen species, decreased NADPH levels, and depleted glutathione as compared to control melanomas. G6PD mutant melanomas compensated for this increase in oxidative stress by increasing malic enzyme activity and glutamine consumption. This generated a new metabolic vulnerability as G6PD mutant melanomas were more dependent upon glutaminase than control melanomas, both for oxidative stress management and anaplerosis. The oxidative pentose phosphate pathway, malic enzyme, and glutaminolysis thus confer layered protection against oxidative stress during metastasis.


Assuntos
Glucosefosfato Desidrogenase/metabolismo , Glutamina/metabolismo , Melanoma/metabolismo , Estresse Oxidativo/fisiologia , Animais , Humanos , Camundongos , Camundongos Endogâmicos NOD , NADP/metabolismo , Oxirredução , Via de Pentose Fosfato/fisiologia , Espécies Reativas de Oxigênio/metabolismo
15.
Exp Dermatol ; 31(5): 700-714, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35030266

RESUMO

The skin's ability to function optimally is affected by many diverse factors. Metabolomics has a great potential to improve our understanding of the underlying metabolic changes and the affected pathways. Therefore, the objective of this study was to review the current state of the literature and to perform further metabolic pathway analysis on the obtained data. The aim was to gain an overview of the metabolic changes under altered conditions and to identify common and different patterns as a function of the investigated factors. A cross-study comparison of the extracted studies from different databases identified 364 metabolites, whose concentrations were considerably altered by the following factor groups: irradiation, xenobiotics, ageing and skin diseases (mainly psoriasis). Using metabolic databases and pathway analysis tools, the individual metabolites were assigned to the corresponding metabolic pathways and the most strongly affected signalling pathways were identified. All factors induced oxidative stress. Thus, antioxidant defence systems, especially coenzyme Q10  (ageing) and the glutathione system (irradiation, ageing, xenobiotics), were impacted. Lipid metabolism was also impacted by all factors studied. The carnitine shuttle as part of ß-oxidation was activated by all factor groups except ageing. Glycolysis, Krebs (TCA) cycle and purine metabolism were mainly affected by irradiation and xenobiotics. The pentose phosphate pathway was activated, and Krebs cycle was downregulated in response to oxidative stress. In summary, it can be ascertained that mainly energy metabolism, lipid metabolism, antioxidative defence and DNA repair systems were impacted by the factors studied.


Assuntos
Metabolômica , Xenobióticos , Ciclo do Ácido Cítrico , Metabolismo Energético , Via de Pentose Fosfato/fisiologia
16.
Physiol Rep ; 9(23): e15137, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34889054

RESUMO

Many of the molecular and cellular mechanisms discovered to regulate skeletal muscle hypertrophy were first identified using the rodent synergist ablation model. This model reveals the intrinsic capability and necessary pathways of skeletal muscle growth in response to mechanical overload (MOV). Reminiscent of the rapid cellular growth observed with cancer, we hypothesized that in response to MOV, skeletal muscle would undergo metabolic programming to sustain increased demands to support hypertrophy. To test this hypothesis, we analyzed the gene expression of specific metabolic pathways taken from transcriptomic microarray data of a MOV time course. We found an upregulation of genes involved in the oxidative branch of the pentose phosphate pathways (PPP) and mitochondrial branch of the folate cycle suggesting an increase in the production of NADPH. In addition, we sought to determine the potential role of skeletal muscle-enriched microRNA (myomiRs) and satellite cells in the regulation of the metabolic pathways that changed during MOV. We observed an inverse pattern in gene expression between muscle-enriched myomiR-1 and its known target gene glucose-6-phosphate dehydrogenase, G6pdx, suggesting myomiR regulation of PPP activation in response to MOV. Satellite cell fusion had a significant but modest impact on PPP gene expression. These transcriptomic findings suggest the robust muscle hypertrophy induced by MOV requires enhanced redox metabolism via PPP production of NADPH which is potentially regulated by a myomiR network.


Assuntos
Hipertrofia/metabolismo , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Doenças Musculares/metabolismo , Via de Pentose Fosfato/fisiologia , Animais , Feminino , Glucosefosfato Desidrogenase/genética , Glucosefosfato Desidrogenase/metabolismo , Glicólise/fisiologia , Hipertrofia/genética , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Doenças Musculares/genética
17.
Front Endocrinol (Lausanne) ; 12: 791174, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34867831

RESUMO

Estrogen therapy is widely used as a supplementary treatment after hysteroscopy for female infertility patients owing to its protective function that improves endometrial regeneration and menstruation, inhibits recurrent adhesions, and improves subsequent conception rate. The endometrial protective function of such estrogen administration pre-surgery is still controversial. In the current study, 12 infertility patients were enrolled, who were treated with estrogen before hysteroscopy surgery. Using cutting-edge metabolomic analysis, we observed alterations in the pentose phosphate pathway (PPP) intermediates of the patient's endometrial tissues. Furthermore, using Ishikawa endometrial cells, we validated our clinical discovery and identified estrogen-ESR-G6PD-PPP axial function, which promotes estrogen-induced cell proliferation.


Assuntos
Proliferação de Células/efeitos dos fármacos , Endométrio/efeitos dos fármacos , Estradiol/uso terapêutico , Estrogênios/uso terapêutico , Metabolômica/métodos , Via de Pentose Fosfato/efeitos dos fármacos , Linhagem Celular Transformada , Proliferação de Células/fisiologia , Endométrio/metabolismo , Estradiol/farmacologia , Estrogênios/farmacologia , Feminino , Humanos , Infertilidade Feminina/tratamento farmacológico , Infertilidade Feminina/metabolismo , Via de Pentose Fosfato/fisiologia
18.
PLoS Biol ; 19(12): e3001468, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34860829

RESUMO

The structure of the metabolic network is highly conserved, but we know little about its evolutionary origins. Key for explaining the early evolution of metabolism is solving a chicken-egg dilemma, which describes that enzymes are made from the very same molecules they produce. The recent discovery of several nonenzymatic reaction sequences that topologically resemble central metabolism has provided experimental support for a "metabolism first" theory, in which at least part of the extant metabolic network emerged on the basis of nonenzymatic reactions. But how could evolution kick-start on the basis of a metal catalyzed reaction sequence, and how could the structure of nonenzymatic reaction sequences be imprinted on the metabolic network to remain conserved for billions of years? We performed an in vitro screening where we add the simplest components of metabolic enzymes, proteinogenic amino acids, to a nonenzymatic, iron-driven reaction network that resembles glycolysis and the pentose phosphate pathway (PPP). We observe that the presence of the amino acids enhanced several of the nonenzymatic reactions. Particular attention was triggered by a reaction that resembles a rate-limiting step in the oxidative PPP. A prebiotically available, proteinogenic amino acid cysteine accelerated the formation of RNA nucleoside precursor ribose-5-phosphate from 6-phosphogluconate. We report that iron and cysteine interact and have additive effects on the reaction rate so that ribose-5-phosphate forms at high specificity under mild, metabolism typical temperature and environmental conditions. We speculate that accelerating effects of amino acids on rate-limiting nonenzymatic reactions could have facilitated a stepwise enzymatization of nonenzymatic reaction sequences, imprinting their structure on the evolving metabolic network.


Assuntos
Cisteína/metabolismo , Ferro/metabolismo , Ribosemonofosfatos/metabolismo , Aminoácidos/metabolismo , Catálise , Cisteína/química , Evolução Molecular , Glucose/metabolismo , Glicólise/fisiologia , Ferro/química , Espectroscopia de Ressonância Magnética/métodos , Redes e Vias Metabólicas/fisiologia , Origem da Vida , Via de Pentose Fosfato/genética , Via de Pentose Fosfato/fisiologia
19.
Cell Rep ; 36(8): 109620, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34433052

RESUMO

Brain function relies almost solely on glucose as an energy substrate. The main model of brain metabolism proposes that glucose is taken up and converted into lactate by astrocytes to fuel the energy-demanding neuronal activity underlying plasticity and memory. Whether direct neuronal glucose uptake is required for memory formation remains elusive. We uncover, in Drosophila, a mechanism of glucose shuttling to neurons from cortex glia, an exclusively perisomatic glial subtype, upon formation of olfactory long-term memory (LTM). In vivo imaging reveals that, downstream of cholinergic activation of cortex glia, autocrine insulin signaling increases glucose concentration in glia. Glucose is then transferred from glia to the neuronal somata in the olfactory memory center to fuel the pentose phosphate pathway and allow LTM formation. In contrast, our results indicate that the increase in neuronal glucose metabolism, although crucial for LTM formation, is not routed to glycolysis.


Assuntos
Memória de Longo Prazo/fisiologia , Neuroglia/metabolismo , Neurônios/metabolismo , Via de Pentose Fosfato/fisiologia , Animais , Astrócitos/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Glucose/metabolismo
20.
Biochem Pharmacol ; 192: 114736, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34411567

RESUMO

Reprogramming of energy metabolism is a hallmarkofcancer, and the pentose phosphate pathway (PPP) is a major glucose metabolic pathway important for meeting the cellular demands of biosynthesis and anti-oxidant defense. Our previous study showed that phosphoinositide 3-kinase enhancer-activating Akt (PIKE-A) plays an important role in glioblastoma cell survival and growth under cellular energy stress condition. However, the crucial functions of PIKE-A in cancer energy metabolism are poorly understood.In the present study, we show that PIKE-A promotes DNA biosynthesis, NADPH production and inhibits reactive oxygen species (ROS) production, leading to increasing proliferation and growth of glioblastoma cell and suppressing cellular senescence. Mechanistically, PIKE-A binds to STAT3 and stimulates its phosphorylation mediated by tyrosine kinase Fyn, which enhances transcription of the rate-limitting enzyme glucose-6-phosphate dehydrogenase (G6PD) in the PPP. Finally, targeting PIKE-A-G6PD axis sensitizes glioblastoma to temozolomide (TMZ)treatment. This study reveals that STAT3 is a novel binding partner of PIKE-A which recruits Fyn to phosphorylate STAT3, contributing to the expression of G6PD, leading to promoting tumor growth and suppressing cellular senescence. Thus, the PIKE-A/STAT3/G6PD axis strongly links the PPP to carcinogenesis and may become a promising cancer therapeutic target.


Assuntos
Proliferação de Células/fisiologia , Proteínas de Ligação ao GTP/biossíntese , Proteínas Ativadoras de GTPase/biossíntese , Glioblastoma/metabolismo , Glucosefosfato Desidrogenase/biossíntese , Via de Pentose Fosfato/fisiologia , Fator de Transcrição STAT3/biossíntese , Animais , Linhagem Celular Tumoral , Regulação Enzimológica da Expressão Gênica , Técnicas de Inativação de Genes/métodos , Glioblastoma/patologia , Glucosefosfato Desidrogenase/genética , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Nus , Fosforilação/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...