Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.974
Filtrer
1.
Phytomedicine ; 134: 155973, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39241384

RÉSUMÉ

BACKGROUND: Inflammatory bowel disease (IBD) is a chronic and relapsing disease marked by chronic tissue inflammation that alters the integrity and function of the gut, seriously impacting patient health and quality of life. Aucklandiae Radix (AR), known as Mu Xiang in Chinese, is a traditional Chinese medicine documented in Chinese Pharmacopoeia with effects of strengthening the intestine and stopping diarrhea. However, the potential of AR in treating intestinal inflammation and its underlying mechanism have yet to be further elucidated. PURPOSE: The objective of this study was to explore the protective effect and the potential mechanism attributable to AR for treating ulcerative colitis (UC). STUDY DESIGN AND METHODS: A murine model of UC was constructed using dextran sulfate sodium (DSS) to examine the therapeutic potential of AR in alleviating inflammation and modulating the immune response. Advanced techniques such as photocrosslinking target fishing technique, click chemistry, Western blot analysis, real-time quantitative PCR, flow cytometry, immunofluorescence, and immunohistochemistry were employed to unveil the therapeutic mechanism of AR for treating IBD. RESULTS: AR decreased disease activity index (DAI) score to alleviate the course of IBD through ameliorating intestinal barrier function in DSS-induced mice. Furthermore, AR suppressed NF-κB and NLRP3 pathways to reduce the release of pro-inflammatory factors interleukin-6 and 1ß (IL-6 and IL-1ß) and tumor necrosis factor α (TNF-α), allowing to alleviate the inflammatory response. Flow cytometry revealed that AR could reduce the accumulation of intestinal macrophages and neutrophils, maintaining intestinal immune balance by regulating the ratio of Treg to Th17 cells. It was worth noting that pyruvate kinase isozyme type M2 (PKM2) served as a potential target of AR using the photocrosslinking target fishing technology, which was further supported by cellular thermal shift assay (CETSA), drug affinity target stability (DARTS), and PKM2 knockdown experiments. CONCLUSION: AR targeted PKM2 to inhibit NF-κB and NLRP3 pathways, thereby modulating the inflammatory response and immunity to alleviate DSS-induced UC. These findings suggested the potential of AR in the treatment of UC and AR as a candidate for developing PKM2 regulators.


Sujet(s)
Rectocolite hémorragique , Sulfate dextran , Modèles animaux de maladie humaine , Médicaments issus de plantes chinoises , Pyruvate kinase , Animaux , Rectocolite hémorragique/traitement médicamenteux , Rectocolite hémorragique/induit chimiquement , Souris , Pyruvate kinase/métabolisme , Médicaments issus de plantes chinoises/pharmacologie , Souris de lignée C57BL , Mâle , Facteur de transcription NF-kappa B/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéines de transport/métabolisme , Protéines membranaires/métabolisme
2.
Physiol Rep ; 12(17): e70040, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39256891

RÉSUMÉ

Cardiac metabolism ensures a continuous ATP supply, primarily using fatty acids in a healthy state and favoring glucose in pathological conditions. Pyruvate kinase muscle (PKM) controls the final step of glycolysis, with PKM1 being the main isoform in the heart. PKM2, elevated in various heart diseases, has been suggested to play a protective role in cardiac stress, but its function in basal cardiac metabolism remains unclear. We examined hearts from global PKM2 knockout (PKM2-/-) mice and found reduced intracellular glucose. Isotopic tracing of U-13C glucose revealed a shift to biosynthetic pathways in PKM2-/- cardiomyocytes. Total ATP content was two-thirds lower in PKM2-/- hearts, and functional analysis indicated reduced mitochondrial oxygen consumption. Total reactive oxygen species (ROS) and mitochondrial superoxide were also increased in PKM2-/- cardiomyocytes. Intriguingly, PKM2-/- hearts had preserved ejection fraction compared to controls. Mechanistically, increased calcium/calmodulin-dependent kinase II activity and phospholamban phosphorylation may contribute to higher sarcoendoplasmic reticulum calcium ATPase 2 pump activity in PKM2-/- hearts. Loss of PKM2 led to altered glucose metabolism, diminished mitochondrial function, and increased ROS in cardiomyocytes. These data suggest that cardiac PKM2 acts as an important rheostat to maintain ATP levels while limiting oxidative stress. Although loss of PKM2 did not impair baseline contractility, its absence may make hearts more sensitive to environmental stress or injury.


Sujet(s)
Myocytes cardiaques , Stress oxydatif , Animaux , Myocytes cardiaques/métabolisme , Souris , Souris knockout , Glucose/métabolisme , Mâle , Espèces réactives de l'oxygène/métabolisme , Protéines de liaison au calcium/métabolisme , Protéines de liaison au calcium/génétique , Mitochondries du myocarde/métabolisme , Sarcoplasmic Reticulum Calcium-Transporting ATPases/métabolisme , Sarcoplasmic Reticulum Calcium-Transporting ATPases/génétique , Souris de lignée C57BL , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique , Adénosine triphosphate/métabolisme , Calcium-Calmodulin-Dependent Protein Kinase Type 2/métabolisme , Calcium-Calmodulin-Dependent Protein Kinase Type 2/génétique , Myocarde/métabolisme
3.
Elife ; 132024 Sep 12.
Article de Anglais | MEDLINE | ID: mdl-39264698

RÉSUMÉ

Reactive astrocytes play critical roles in the occurrence of various neurological diseases such as multiple sclerosis. Activation of astrocytes is often accompanied by a glycolysis-dominant metabolic switch. However, the role and molecular mechanism of metabolic reprogramming in activation of astrocytes have not been clarified. Here, we found that PKM2, a rate-limiting enzyme of glycolysis, displayed nuclear translocation in astrocytes of EAE (experimental autoimmune encephalomyelitis) mice, an animal model of multiple sclerosis. Prevention of PKM2 nuclear import by DASA-58 significantly reduced the activation of mice primary astrocytes, which was observed by decreased proliferation, glycolysis and secretion of inflammatory cytokines. Most importantly, we identified the ubiquitination-mediated regulation of PKM2 nuclear import by ubiquitin ligase TRIM21. TRIM21 interacted with PKM2, promoted its nuclear translocation and stimulated its nuclear activity to phosphorylate STAT3, NF-κB and interact with c-myc. Further single-cell RNA sequencing and immunofluorescence staining demonstrated that TRIM21 expression was upregulated in astrocytes of EAE. TRIM21 overexpressing in mice primary astrocytes enhanced PKM2-dependent glycolysis and proliferation, which could be reversed by DASA-58. Moreover, intracerebroventricular injection of a lentiviral vector to knockdown TRIM21 in astrocytes or intraperitoneal injection of TEPP-46, which inhibit the nuclear translocation of PKM2, effectively decreased disease severity, CNS inflammation and demyelination in EAE. Collectively, our study provides novel insights into the pathological function of nuclear glycolytic enzyme PKM2 and ubiquitination-mediated regulatory mechanism that are involved in astrocyte activation. Targeting this axis may be a potential therapeutic strategy for the treatment of astrocyte-involved neurological disease.


Sujet(s)
Astrocytes , Encéphalomyélite auto-immune expérimentale , Ribonucléoprotéines , Régulation positive , Animaux , Astrocytes/métabolisme , Encéphalomyélite auto-immune expérimentale/métabolisme , Encéphalomyélite auto-immune expérimentale/génétique , Souris , Ribonucléoprotéines/métabolisme , Ribonucléoprotéines/génétique , Hormones thyroïdiennes/métabolisme , Hormones thyroïdiennes/génétique , , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Souris de lignée C57BL , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique , Transport nucléaire actif , Femelle , Glycolyse , Ubiquitination , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Noyau de la cellule/métabolisme
4.
Expert Opin Ther Pat ; 34(10): 953-962, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39279560

RÉSUMÉ

INTRODUCTION: Cancer cells adopt a glycolytic phenotype to fulfill their energy needs in unfavorable conditions. In metabolic rewiring, cancer cells upregulate the expression of glycolytic pathway regulators including glucose transporter 1, hexokinase 2, and PKM2 (pyruvate kinase) into its M2 splice form. Among these regulators, PKM2 plays a major role in metabolic reprogramming and is overexpressed in various diseases, including cancer. Dimerization of PKM2 causes the generation of synthetic precursors from glycolytic intermediates, which are essential for cellular growth and cancer cell proliferation. COVERED AREAS: This article is focused on examining recent patents (2018-2023) on PKM2 activators, inhibitors and their biological and synthesis properties by using the advanced search service of the European Patent Office (EPO). Moreover, other databases including PubMed, Google Scholar and Elsevier were also examined for scientific data. On basis of their chemical structures, PKM2 activators and inhibitors are classified into pyrazole, pyrolidine-pyrazole, phenol, benzoxazine, isoselenazolo-pyridinium, phthalazine, and propiolylamide derivatives. EXPERT OPINION: Activating PKM2 reduces proliferation and development of cells by reducing the quantity of biomolecules needed for cell formation. PKM2 activators and inhibitors are highly effective in treating many cancer pathogens. It is important to find new, more potent and selective molecules for PKM2 activation and inhibition.


Sujet(s)
Antinéoplasiques , Prolifération cellulaire , Tumeurs , Brevets comme sujet , Pyruvate kinase , Humains , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Animaux , Antinéoplasiques/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Pyruvate kinase/métabolisme , Pyruvate kinase/antagonistes et inhibiteurs , , Protéines membranaires/métabolisme , Protéines membranaires/antagonistes et inhibiteurs , Thérapie moléculaire ciblée , Hormones thyroïdiennes/métabolisme , Protéines de transport/métabolisme , Protéines de transport/antagonistes et inhibiteurs , Développement de médicament , Glycolyse/effets des médicaments et des substances chimiques
5.
Mol Med ; 30(1): 146, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39261768

RÉSUMÉ

BACKGROUND: At present, hepatic ischemia-reperfusion injury (IRI) is an important complication of partial hepatectomy and liver transplantation, and it is an important cause of poor prognosis. Spleen tyrosine kinase(SYK) plays an important role in a variety of signaling pathways in the liver, but its role in hepatic IRI is still unclear. This study aims to investigate the role and mechanism of SYK in hepatic IRI and tumor recurrence. METHODS: We first observed the activation of SYK in the liver of mice in response to hepatic IRI. Subsequently, Pharmacological inhibitions of SYK were used to evaluated the effect of SYK on neutrophil recruitment and NETosis, and further explored the effect of SYK on IRI and tumor recurrence. RESULTS: Our study shows that SYK is activated in response to hepatic IRI and aggravates liver injury. On the one hand, neutrophils SYK during the early stage of liver reperfusion increases neutrophil extracellular traps (NETs) production by promoting Pyruvate kinase M2(PKM2) nuclear translocation leading to upregulation of phosphorylated STAT3, thereby exacerbating liver inflammation and tumor recurrence. On the other hand, macrophages SYK can promote the recruitment of neutrophils and increase the activation of NLRP3 inflammasome and IL1ß, which further promotes the formation of NETs. CONCLUSIONS: Our study demonstrates that neutrophil and macrophage SYK synergistically promote hepatic IRI and tumor recurrence, and SYK may be a potential target to improve postoperative hepatic IRI and tumor recurrence.


Sujet(s)
Pièges extracellulaires , Protéines membranaires , Granulocytes neutrophiles , Lésion d'ischémie-reperfusion , Facteur de transcription STAT-3 , Syk kinase , Syk kinase/métabolisme , Animaux , Facteur de transcription STAT-3/métabolisme , Pièges extracellulaires/métabolisme , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/anatomopathologie , Phosphorylation , Souris , Protéines membranaires/métabolisme , Mâle , Granulocytes neutrophiles/métabolisme , Protéines de transport/métabolisme , Pyruvate kinase/métabolisme , Foie/métabolisme , Foie/anatomopathologie , , Récidive tumorale locale/métabolisme , Souris de lignée C57BL , Modèles animaux de maladie humaine , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Humains , Transduction du signal
6.
Dev Biol ; 516: 122-129, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39117030

RÉSUMÉ

Growing evidence suggests that metabolic regulation directly influences cellular function and development and thus may be more dynamic than previously expected. In vivo and in real-time analysis of metabolite activities during development is crucial to test this idea directly. In this study, we employ two metabolic biosensors to track the dynamics of pyruvate and oxidative phosphorylation (Oxphos) during the early embryogenesis of the sea urchin. A pyruvate sensor, PyronicSF, shows the signal enrichment on the mitotic apparatus, which is consistent with the localization patterns of the corresponding enzyme, pyruvate kinase (PKM). The addition of pyruvate increases the PyronicSF signal, while PKM knockdown decreases its signal, responding to the pyruvate level in the cell. Similarly, a ratio-metric sensor, Grx-roGFP, that reads the redox potential of the cell responds to DTT and H2O2, the known reducer and inducer of Oxphos. These observations suggest that these metabolic biosensors faithfully reflect the metabolic status in the cell during embryogenesis. The time-lapse imaging of these biosensors suggests that pyruvate and Oxphos levels change both spatially and temporarily during embryonic development. Pyruvate level is increased first in micromeres compared to other blastomeres at the 16-cell stage and remains high in ectoderm while decreasing in endomesoderm during gastrulation. In contrast, the Oxphos signal first decreases in micromeres at the 16-cell stage, while it increases in the endomesoderm during gastrulation, showing the opposite trend of the pyruvate signal. These results suggest that metabolic regulation is indeed both temporally and spatially dynamic during embryogenesis, and these biosensors are a valuable tool to monitor metabolic activities in real-time in developing embryos.


Sujet(s)
Techniques de biocapteur , Développement embryonnaire , Phosphorylation oxydative , Pyruvate kinase , Acide pyruvique , Echinoidea , Animaux , Techniques de biocapteur/méthodes , Acide pyruvique/métabolisme , Pyruvate kinase/métabolisme , Echinoidea/embryologie , Echinoidea/métabolisme , Embryon non mammalien/métabolisme , Imagerie accélérée/méthodes
7.
Int J Chron Obstruct Pulmon Dis ; 19: 1905-1920, 2024.
Article de Anglais | MEDLINE | ID: mdl-39206144

RÉSUMÉ

Purpose: The main objective of this study was to explore the mechanism of effective component compatibility of Bufei Yishen formula III (ECC-BYF III) in inhibiting mitochondrial oxidative stress in a rat model of chronic obstructive pulmonary disease (COPD). Methods: A549 cells exposed to cigarette smoke extract (CSE) were used to establish a model of mitochondrial oxidative damage. The cells were treated with the plasmid encoding Pkm2 and the enzymes and proteins involved in oxidative stress and mitochondrial function were measured. A rat model of COPD was established using CS and bacteria. Two different treatments were established, ECC-BYF III (5.5 mg/kg/d) and N-acetylcysteine (54 mg/kg/day). Animals were tested for pulmonary function (Vt, PEF, FVC, FEV0.1s and Cdyn) after eight weeks of therapy and were sacrificed. Pulmonary H&E staining was performed, and the total superoxide dismutase (T-SOD), glutathione peroxidase (GSH-Px), total antioxidant capacity (T-AOC), and malondialdehyde (MDA) content were measured. The mitochondrial function was also examined. Furthermore, the Pkm2/Nrf2 signaling pathway was evaluated. Results: Overexpression of Pkm2 dramatically ameliorated the CS-induced mitochondrial oxidative damage. Further studies indicated that ECC-BYF III significantly improved mitochondrial function and inhibited oxidative stress in the lung tissues of COPD rats. Moreover, it can upregulate mitochondrial respiratory chain enzyme activity. ECC-BYF III also decreased the MDA content and increased T-SOD, GSH-Px, and T-AOC expression to facilitate oxidative homeostasis. Finally, our results indicated that the Pkm2/Nrf2 pathway is regulated by ECC-BYF III in A549 cells and lung tissue. Conclusion: These results indicate that ECC-BYF III exerts a strong effective therapeutic effect against cigarette smoke combined with bacteria-induced COPD in rats by activating the Pkm2/Nrf2 signaling pathway and restoring mitochondrial oxidative stress. Although more in vivo animal model research is needed to confirm these findings, this study contributes new data to support the conventional usage of ECC-BYF III.


Sujet(s)
Modèles animaux de maladie humaine , Médicaments issus de plantes chinoises , Poumon , Mitochondries , Facteur-2 apparenté à NF-E2 , Stress oxydatif , Broncho-pneumopathie chronique obstructive , Transduction du signal , Animaux , Facteur-2 apparenté à NF-E2/métabolisme , Broncho-pneumopathie chronique obstructive/traitement médicamenteux , Broncho-pneumopathie chronique obstructive/métabolisme , Broncho-pneumopathie chronique obstructive/physiopathologie , Humains , Stress oxydatif/effets des médicaments et des substances chimiques , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Poumon/physiopathologie , Poumon/enzymologie , Médicaments issus de plantes chinoises/pharmacologie , Cellules A549 , Mâle , , Pyruvate kinase/métabolisme , Antioxydants/pharmacologie , Rat Sprague-Dawley , Protéines de transport/métabolisme , Hormones thyroïdiennes/métabolisme , Fumée/effets indésirables , Protéines membranaires/métabolisme , Protéines membranaires/génétique
8.
Int Immunopharmacol ; 141: 112880, 2024 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-39153304

RÉSUMÉ

Pyruvate kinase M2 (PKM2) is a key metabolic enzyme. Yet, its role in cerebral ischemia injury remains unclear. In this study we demonstrated that PKM2 expression was increased in the microglia after mouse cerebral ischemia-reperfusion (I/R) injury. We found that microglial polarization-mediated pro-inflammatory effect was mediated by PKM2 after cerebral I/R. Mechanistically, our results revealed that nuclear PKM2 mediated ischemia-induced microglial polarization through association with acetyl-H3K9. Hif-1α mediated the effect of nuclear PKM2/histone H3 on microglial polarization. PKM2-dependent Histone H3/Hif-1α modifications contributed the expression of CCL2 and induced up-regulation of microglial polarization in peri-infarct, resulting in neuroinflammation. Inhibiting nuclear translocation of microglial PKM2 reduced ischemia-induced pro-inflammation and promoted neuronal survival. Together, this study identifies nucleus PKM2 as a crucial mediator for regulating ischemia-induced neuroinflammation, suggesting PKM2 as a potential therapeutic target in ischemic stroke.


Sujet(s)
Souris de lignée C57BL , Microglie , Maladies neuro-inflammatoires , Pyruvate kinase , Lésion d'ischémie-reperfusion , Animaux , Microglie/métabolisme , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/anatomopathologie , Mâle , Pyruvate kinase/métabolisme , Souris , Maladies neuro-inflammatoires/anatomopathologie , Maladies neuro-inflammatoires/immunologie , Maladies neuro-inflammatoires/métabolisme , Encéphalopathie ischémique/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Régulation négative , Noyau de la cellule/métabolisme , Histone/métabolisme , Neuroprotection , Chimiokine CCL2/métabolisme , Chimiokine CCL2/génétique , Cellules cultivées , Modèles animaux de maladie humaine , Neurones/métabolisme , Neurones/anatomopathologie , Hormones thyroïdiennes/métabolisme
9.
Nat Commun ; 15(1): 7463, 2024 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-39198443

RÉSUMÉ

Most cancer cells reprogram their glucose metabolic pathway from oxidative phosphorylation to aerobic glycolysis for energy production. By reducing enzyme activity of pyruvate kinase M2 (PKM2), cancer cells attain a greater fraction of glycolytic metabolites for macromolecule synthesis needed for rapid proliferation. Here we demonstrate that hydrogen sulfide (H2S) destabilizes the PKM2 tetramer into monomer/dimer through sulfhydration at cysteines, notably at C326, leading to reduced PKM2 enzyme activity and increased PKM2-mediated transcriptional activation. Blocking PKM2 sulfhydration at C326 through amino acid mutation stabilizes the PKM2 tetramer and crystal structure further revealing the tetramer organization of PKM2-C326S. The PKM2-C326S mutant in cancer cells rewires glucose metabolism to mitochondrial respiration, significantly inhibiting tumor growth. In this work, we demonstrate that PKM2 sulfhydration by H2S inactivates PKM2 activity to promote tumorigenesis and inhibiting this process could be a potential therapeutic approach for targeting cancer metabolism.


Sujet(s)
Glucose , Sulfure d'hydrogène , Sulfure d'hydrogène/métabolisme , Humains , Glucose/métabolisme , Animaux , Lignée cellulaire tumorale , Souris , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique , Pyruvate kinase/composition chimique , Cystéine/métabolisme , Glycolyse , Hormones thyroïdiennes/métabolisme , Mutation , Mitochondries/métabolisme , Tumeurs/métabolisme , Tumeurs/génétique , Tumeurs/anatomopathologie , Multimérisation de protéines , Souris nude , Protéines de transport/métabolisme , Protéines de transport/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique ,
10.
Mol Med Rep ; 30(4)2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39155878

RÉSUMÉ

Glycolysis occurs in all living organisms as a form of energy supply. Pyruvate kinase M2 (PKM2) is one of the rate­limiting enzymes in the glycolytic process. PKM2 is considered to serve an important role in several terminal diseases, including sepsis. However, to the best of our knowledge, the specific mechanistic role of PKM2 in sepsis remains to be systematically summarised. Therefore, the present review aims to summarise the roles of PKM2 in sepsis progression. In addition, potential treatment strategies for patients with sepsis are discussed. The present review hopes to lay the groundwork for studying the role of PKM2 and developing therapeutic strategies against metabolic disorders that occur during sepsis.


Sujet(s)
Pyruvate kinase , Sepsie , Humains , Sepsie/métabolisme , Pyruvate kinase/métabolisme , Glycolyse , Animaux , , Hormones thyroïdiennes/métabolisme
11.
J Agric Food Chem ; 72(32): 18013-18026, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39088205

RÉSUMÉ

Glucose and lipid metabolism dysregulation in skeletal muscle contributes to the development of metabolic disorders. The efficacy of fucoxanthin in alleviating lipid metabolic disorders in skeletal muscle remains poorly understood. In this study, we systematically investigated the impact of fucoxanthin on mitigating lipid deposition and insulin resistance in skeletal muscle employing palmitic acid-induced lipid deposition in C2C12 cells and ob/ob mice. Fucoxanthin significantly alleviated PA-induced skeletal muscle lipid deposition and insulin resistance. In addition, fucoxanthin prominently upregulated the expression of lipid metabolism-related genes (Pparα and Cpt-1), promoting fatty acid ß-oxidation metabolism. Additionally, fucoxanthin significantly increased the expression of Pgc-1α and Tfam, elevated the mtDNA/nDNA ratio, and reduced ROS levels. Further, we identified pyruvate kinase muscle isozyme 1 (PKM1) as a high-affinity protein for fucoxanthin by drug affinity-responsive target stability and LC-MS and confirmed their robust interaction by CETSA, microscale thermophoresis, and circular dichroism. Supplementation with pyruvate, the product of PKM1, significantly attenuated the beneficial effects of fucoxanthin on lipid deposition and insulin resistance. Mechanistically, fucoxanthin reduced glucose glycolysis rate and enhanced mitochondrial biosynthesis and fatty acid ß-oxidation through inhibiting PKM1 activity, thereby alleviating lipid metabolic stress. These findings present a novel clinical strategy for treating metabolic diseases using fucoxanthin.


Sujet(s)
Insulinorésistance , Métabolisme lipidique , Muscles squelettiques , Pyruvate kinase , Xanthophylles , Animaux , Souris , Muscles squelettiques/métabolisme , Muscles squelettiques/effets des médicaments et des substances chimiques , Xanthophylles/pharmacologie , Métabolisme lipidique/effets des médicaments et des substances chimiques , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique , Mâle , Humains , Souris de lignée C57BL , Alimentation riche en graisse/effets indésirables
12.
J Neuroimmune Pharmacol ; 19(1): 48, 2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39196455

RÉSUMÉ

Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. Microglial activation and neuroinflammation are key cellular events that determine the outcome of TBI, especially neuronal and cognitive function. Studies have suggested that the metabolic characteristics of microglia dictate their inflammatory response. The pyruvate kinase isoform M2 (PKM2), a key glycolytic enzyme, is involved in the regulation of various cellular metabolic processes, including mitochondrial metabolism. This suggests that PKM2 may also participate in the regulation of microglial activation during TBI. Therefore, the present study aimed to evaluate the role of PKM2 in regulating microglial activation and neuroinflammation and its effects on cognitive function following TBI. A controlled cortical impact (CCI) mouse model and inflammation-induced primary mouse microglial cells in vitro were used to investigate the potential effects of PKM2 inhibition and regulation. PKM2 was significantly increased during the acute and subacute phases of TBI and was predominantly detected in microglia rather than in neurons. Our results demonstrate that shikonin and TEPP-46 can inhibit microglial inflammation, improving mitochondria, improving mouse behavior, reducing brain defect volume, and alleviating pathological changes after TBI. There is a difference in the intervention of shikonin and TEPP-46 on PKM2. Shikonin directly inhibits General PKM2; TEPP-46 can promote the expression of PKM2 tetramer. In vitro experiments, TEPP-46 can promote the expression of PKM2 tetramer, enhance the interaction between PKM2 and MFN2, improve mitochondria, alleviate neuroinflammation. General inhibition and tetramerization activation of PKM2 attenuated cognitive function caused by TBI, whereas PKM2 tetramerization exhibited a better treatment effect. Our experiments demonstrated the non-metabolic role of PKM2 in the regulation of microglial activation following TBI. Both shikonin and TEPP-46 can inhibit pro-inflammatory factors, but only TEPP-46 can promote PKM2 tetramerization and upregulate the release of anti-inflammatory factors from microglia.


Sujet(s)
Lésions traumatiques de l'encéphale , Microglie , Mitochondries , Naphtoquinones , Pyruvate kinase , Animaux , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Lésions traumatiques de l'encéphale/métabolisme , Lésions traumatiques de l'encéphale/anatomopathologie , Lésions traumatiques de l'encéphale/traitement médicamenteux , Souris , Pyruvate kinase/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Naphtoquinones/pharmacologie , Mâle , Souris de lignée C57BL , Multimérisation de protéines/effets des médicaments et des substances chimiques , Cellules cultivées
13.
J Biochem Mol Toxicol ; 38(8): e23799, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39132768

RÉSUMÉ

It is well established that pyruvate kinase M2 (PKM2) activity contributes to metabolic reprogramming in various cancers, including colorectal cancer (CRC). Estrogen or 17ß-estradiol (E2) signaling is also known to modulate glycolysis markers in cancer cells. However, whether the inhibition of PKM2 combined with E2 treatment could adversely affect glucose metabolism in CRC cells remains to be investigated. First, we confirmed the metabolic plasticity of CRC cells under varying environmental conditions. Next, we identified glycolysis markers that were upregulated in CRC patients and assessed in vitro mRNA levels following E2 treatment. We found that PKM2 expression, which is highly upregulated in CRC clinical samples, is not altered by E2 treatment in CRC cells. In this study, glucose uptake, generation of reactive oxygen species (ROS), lactate production, cell viability, and apoptosis were evaluated in CRC cells following E2 treatment, PKM2 silencing, or a combination of both. Compared to individual treatments, combination therapy resulted in a significant reduction in cell viability and enhanced apoptosis. Glucose uptake and ROS production were markedly reduced in PKM2-silenced E2-treated cells. The data presented here suggest that E2 signaling combined with PKM2 inhibition cumulatively targets glucose metabolism in a manner that negatively impacts CRC cell growth. These findings hold promise for novel therapeutic strategies targeting altered metabolic pathways in CRC.


Sujet(s)
Tumeurs colorectales , Humains , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/métabolisme , Tumeurs colorectales/génétique , Hormones thyroïdiennes/métabolisme , Lignée cellulaire tumorale , Espèces réactives de l'oxygène/métabolisme , Oestrogènes/pharmacologie , , Oestradiol/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Glucose/métabolisme , Protéines de transport/métabolisme , Pyruvate kinase/métabolisme , Pyruvate kinase/antagonistes et inhibiteurs , Pyruvate kinase/génétique , Glycolyse/effets des médicaments et des substances chimiques , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Femelle
14.
Cell Rep Med ; 5(8): 101684, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39128469

RÉSUMÉ

Sirtuin 1 (SIRT1) is a histone deacetylase and plays diverse functions in various physiological events, from development to lifespan regulation. Here, in Parkinson's disease (PD) model mice, we demonstrated that SIRT1 ameliorates parkinsonism, while SIRT1 knockdown further aggravates PD phenotypes. Mechanistically, SIRT1 interacts with and deacetylates pyruvate kinase M2 (PKM2) at K135 and K206, thus leading to reduced PKM2 enzyme activity and lactate production, which eventually results in decreased glial activation in the brain. Administration of lactate in the brain recapitulates PD-like phenotypes. Furthermore, increased expression of PKM2 worsens PD symptoms, and, on the contrary, inhibition of PKM2 by shikonin or PKM2-IN-1 alleviates parkinsonism in mice. Collectively, our data indicate that excessive lactate in the brain might be involved in the progression of PD. By improving lactate homeostasis, SIRT1, together with PKM2, are likely drug targets for developing agents for the treatment of neurodegeneration in PD.


Sujet(s)
Encéphale , Homéostasie , Acide lactique , Pyruvate kinase , Sirtuine-1 , Sirtuine-1/métabolisme , Sirtuine-1/génétique , Animaux , Encéphale/métabolisme , Encéphale/anatomopathologie , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique , Souris , Acide lactique/métabolisme , Humains , Acétylation/effets des médicaments et des substances chimiques , Syndromes parkinsoniens/métabolisme , Syndromes parkinsoniens/anatomopathologie , Syndromes parkinsoniens/traitement médicamenteux , Syndromes parkinsoniens/génétique , Modèles animaux de maladie humaine , Mâle , Souris de lignée C57BL , , Hormones thyroïdiennes/métabolisme , Naphtoquinones/pharmacologie
15.
J Ethnopharmacol ; 335: 118679, 2024 Dec 05.
Article de Anglais | MEDLINE | ID: mdl-39121930

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Sanqi oral solution (SQ) is a traditional Chinese patent medicine, widely used to treat chronic kidney diseases (CKD) in the clinic in China. Previous studies have confirmed its anti-renal fibrosis effect, but the specific pharmacological mechanism is still unclear. AIM OF THE STUDY: Focusing on energy metabolism in fibroblasts, the renoprotective mechanism of SQ was investigated in vitro and in vivo. METHODS: Firstly, the fingerprint of SQ was constructed and its elementary chemical composition was analyzed. In the 5/6Nx rats experiment, the efficacy of SQ on the kidney was evaluated by detecting serum and urine biochemical indexes and pathological staining of renal tissues. Lactic acid and pyruvic acid levels in serum and renal tissues were detected. PCNA protein expression in kidney tissue was detected by immunofluorescence assay and Western blot. Expression levels of HIF-1α, PKM2 and HK2 were determined by immunohistochemistry, Western blot or RT-qPCR assay. In addition, the effect of SQ intervention on cell proliferation and glycolysis was evaluated in TGF-ß1-induced NRK-49F cells, and the role of SQ exposure and HIF-1α/PKM2/glycolysis pathway were further investigated by silencing and overexpressing HIF-1α gene in NRK-49F cells. RESULTS: In 5/6 Nx rats, SQ effectively improved renal function and treated renal injury. It reduced the levels of lactic acid and pyruvic acid in kidney homogenates from CKD rats and decreased the expression levels of HIF-1α, PKM2, HK2, α-SMA, vimentin, collagen I and PCNA in kidney tissues. Similar results were observed in vitro. SQ inhibited NRK-49F cell proliferation, glycolysis and the expression levels of HIF-1α, PKM2 induced by TGF-ß1. Furthermore, we established NRK-49F cells transfected with siRNA or pDNA to silence or overexpress the HIF-1α gene. Overexpression of HIF-1α promoted cellular secretion of lactic acid and pyruvic acid in TGF-ß1-induced NRK-49F cells, however, this change was reversed by intervention with SQ or silencing the HIF-1α gene. Overexpression of HIF-1α can further induce increased PKM2 expression, while SQ intervention can reduce PKM2 expression. Moreover, PKM2 expression was also inhibited after silencing HIF-1α gene, and SQ was not effective even when given. CONCLUSION: The mechanism of action of SQ was explored from the perspective of energy metabolism, and it was found to regulate PKM2-activated glycolysis, inhibit fibroblast activation, and further ameliorate renal fibrosis in CKD by targeting HIF-1α.


Sujet(s)
Fibroblastes , Fibrose , Glycolyse , Sous-unité alpha du facteur-1 induit par l'hypoxie , Rein , Rat Sprague-Dawley , Insuffisance rénale chronique , , Animaux , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Insuffisance rénale chronique/traitement médicamenteux , Insuffisance rénale chronique/métabolisme , Fibrose/traitement médicamenteux , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Mâle , Glycolyse/effets des médicaments et des substances chimiques , Rats , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Rein/métabolisme , Médicaments issus de plantes chinoises/pharmacologie , Lignée cellulaire , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Hormones thyroïdiennes , Administration par voie orale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Protéines de transport/métabolisme , Protéines de transport/génétique , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/génétique
16.
J Coll Physicians Surg Pak ; 34(7): 811-816, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38978246

RÉSUMÉ

OBJECTIVE: To investigate the expression level of pyruvate kinase M1 (PKM1) in patients with acute myeloid leukaemia (AML) as well as its clinical significance. STUDY DESIGN: A case-control study. Place and Duration of the Study: Department of Haematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China, from January 2013 to 2023. METHODOLOGY: The expression levels of PKM1 and pyruvate kinase m2 (PKM2) in the bone marrow of 65 AML patients (excluding M3) and 31 healthy volunteers were determined using reverse transcription-quantitative polymerase chain reaction (RT-qPCR), a method that measures fluorescence in real-time. The associations between PKM1, PKM2 expressions, clinical parameters, and the survival and prognosis of AML patients were analysed. RESULTS: AML patients showed higher PKM1 expression compared to controls. The area under the curve (AUC) of the receiver operating characteristics (ROC) was 0.65 (p = 0.017). PKM1 expression was correlated with peripheral blood leukocyte count (r = -0.276, p = 0.026), CCAAT enhancer-binding protein alpha CEBPA mutation (r = -0.306, p = 0.014), and chemotherapy-induced response (r = -0.292, p = 0.018). Patients with high PKM1 expression had a lower remission rate (p = 0.019) and long-term survival rate (p = 0.034) than those with low PKM1 expression. Patients with AML showed a rise in PKM2 levels; however, the variation was not statistically significant (p >0.05). CONCLUSION: PKM1 expression is upregulated in AML and patients with high PKM1 expression have a lower survival rate. KEY WORDS: PKM1, Acute myeloid leukaemia, Clinical prognosis.


Sujet(s)
Protéines de transport , Leucémie aigüe myéloïde , Protéines membranaires , , Hormones thyroïdiennes , Adulte , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Jeune adulte , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Protéines de transport/génétique , Protéines de transport/métabolisme , Études cas-témoins , Chine/épidémiologie , Leucémie aigüe myéloïde/génétique , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Pronostic , Pyruvate kinase/génétique , Pyruvate kinase/métabolisme , Hormones thyroïdiennes/sang , Hormones thyroïdiennes/métabolisme
17.
Bioorg Med Chem Lett ; 110: 129865, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-38950758

RÉSUMÉ

Pyruvate kinase (PK) is an essential component of cellular metabolism, converting ADP and phosphoenolpyruvate (PEP) to pyruvate in the final step of glycolysis. Of the four unique isoforms of pyruvate kinase, R (PKR) is expressed exclusively in red blood cells and is a tetrameric enzyme that depends on fructose-1,6-bisphosphate (FBP) for activation. PKR deficiency leads to hemolysis of red blood cells resulting in anemia. Activation of PKR in both sickle cell disease and beta-thalassemia patients could lead to improved red blood cell fitness and survival. The discovery of a novel series of substituted urea PKR activators, via the serendipitous identification and diligent characterization of a minor impurity in an High Throughput Screening (HTS) hit will be discussed.


Sujet(s)
Tests de criblage à haut débit , Pyruvate kinase , Pyruvate kinase/métabolisme , Pyruvate kinase/antagonistes et inhibiteurs , Humains , Découverte de médicament , Relation structure-activité , Urée/composition chimique , Urée/pharmacologie , Activateurs d'enzymes/pharmacologie , Activateurs d'enzymes/composition chimique , Activateurs d'enzymes/synthèse chimique , Structure moléculaire , Animaux
18.
Biochim Biophys Acta Mol Cell Res ; 1871(7): 119796, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39038610

RÉSUMÉ

Pyruvate kinase M2 (PKM2) is a key glycolytic enzyme interacting with the inositol 1,4,5-trisphosphate receptor (IP3R). This interaction suppresses IP3R-mediated cytosolic [Ca2+] rises. As PKM2 exists in monomeric, dimeric and tetrameric forms displaying different properties including catalytic activity, we investigated the molecular determinants of PKM2 enabling its interaction with IP3Rs. Treatment of HeLa cells with TEPP-46, a compound stabilizing the tetrameric form of PKM2, increased both its catalytic activity and the suppression of IP3R-mediated Ca2+ signals. Consistently, in PKM2 knock-out HeLa cells, PKM2C424L, a tetrameric, highly active PKM2 mutant, but not inactive PKM2K270M or the less active PKM2K305Q, suppressed IP3R-mediated Ca2+ release. Surprisingly, however, in vitro assays did not reveal a direct interaction between purified PKM2 and either the purified Fragment 5 of IP3R1 (a.a. 1932-2216) or the therein located D5SD peptide (a.a. 2078-2098 of IP3R1), the presumed interaction sites of PKM2 on the IP3R. Moreover, on-nucleus patch clamp of heterologously expressed IP3R1 in DT40 cells devoid of endogenous IP3Rs did not reveal any functional effect of purified wild-type PKM2, mutant PKM2 or PKM1 proteins. These results indicate that an additional factor mediates the regulation of the IP3R by PKM2 in cellulo. Immunoprecipitation of GRP75 using HeLa cell lysates co-precipitated IP3R1, IP3R3 and PKM2. Moreover, the D5SD peptide not only disrupted PKM2:IP3R, but also PKM2:GRP75 and GRP75:IP3R interactions. Our data therefore support a model in which catalytically active, tetrameric PKM2 suppresses Ca2+ signaling via the IP3R through a multiprotein complex involving GRP75.


Sujet(s)
Récepteurs à l'inositol 1,4,5-triphosphate , Protéines membranaires , Humains , Récepteurs à l'inositol 1,4,5-triphosphate/métabolisme , Récepteurs à l'inositol 1,4,5-triphosphate/génétique , Cellules HeLa , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique , Signalisation calcique , Protéines du choc thermique HSP70/métabolisme , Protéines du choc thermique HSP70/génétique , Calcium/métabolisme , Liaison aux protéines , Multimérisation de protéines
19.
Nat Commun ; 15(1): 5761, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982055

RÉSUMÉ

While protein aggregation's association with aging and age-related diseases is well-established, the specific proteins involved and whether dissolving them could alleviate aging remain unclear. Our research addresses this gap by uncovering the role of PKM2 aggregates in aging. We find that PKM2 forms aggregates in senescent cells and organs from aged mice, impairing its enzymatic activity and glycolytic flux, thereby driving cells into senescence. Through a rigorous two-step small molecule library screening, we identify two compounds, K35 and its analog K27, capable of dissolving PKM2 aggregates and alleviating senescence. Further experiments show that treatment with K35 and K27 not only alleviate aging-associated signatures but also extend the lifespan of naturally and prematurely aged mice. These findings provide compelling evidence for the involvement of PKM2 aggregates in inducing cellular senescence and aging phenotypes, and suggest that targeting these aggregates could be a promising strategy for anti-aging drug discovery.


Sujet(s)
Vieillissement , Vieillissement de la cellule , , Animaux , Vieillissement/métabolisme , Souris , Humains , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Protéines de transport/métabolisme , Glycolyse , Hormones thyroïdiennes/métabolisme , Agrégats de protéines , Pyruvate kinase/métabolisme , Souris de lignée C57BL , Mâle
20.
Chem Res Toxicol ; 37(8): 1394-1403, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39066737

RÉSUMÉ

Aerobic glycolysis is a metabolic reprogramming of tumor cells that is essential for sustaining their phenotype of fast multiplication by continuously supplying energy and mass. Pyruvate kinase M2 (PKM2) has a vital role in this process, which has given it high interest as a target for anticancer drug development. With potent toxicity to many types of cancer cells, polyphyllin II (PP2), a steroidal saponin isolated from the herbaceous plant Rhizoma paridis, brought to our attention that it might interfere with the PKM2 activity. In this study, we discovered that PP2 was a novel agonist of PKM2. PP2 activated recombinant PKM2 and changed the protein's oligomeric state to activate intracellular PKM2. At the same time, PP2 suppressed its protein kinase function by decreasing the content of nuclear PKM2. The mRNA levels of its downstream genes, such as Glut1, LDHA, and MYC, were inhibited. In addition, PP2 induced oxidative stress by downregulating the expression and activity of antioxidant proteins such as NQO1, TrxR, and Trx in HT-1080 cells, which in turn led to mitochondrial dysfunction and ultimately induced apoptosis. Moreover, PP2 reduced the proliferation and migration of HT-1080 cells. Thus, targeting the glycolysis pathway offers an unprecedented mode of action for comprehending PP2's pharmacological impacts and advances PP2's further development in fibrosarcoma therapy.


Sujet(s)
Apoptose , Pyruvate kinase , Saponines , Lignée cellulaire tumorale , Pyruvate kinase/antagonistes et inhibiteurs , Pyruvate kinase/génétique , Pyruvate kinase/métabolisme , Saponines/pharmacologie , Saponines/toxicité , Activation enzymatique/effets des médicaments et des substances chimiques , Homéostasie/effets des médicaments et des substances chimiques , Mitochondries/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Fibrosarcome , Humains
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE