Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.126
Filtrer
1.
Cell Mol Life Sci ; 81(1): 254, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38856931

RÉSUMÉ

The endogenous mitochondrial quality control (MQC) system serves to protect mitochondria against cellular stressors. Although mitochondrial dysfunction contributes to cardiac damage during many pathological conditions, the regulatory signals influencing MQC disruption during septic cardiomyopathy (SC) remain unclear. This study aimed to investigate the involvement of pyruvate kinase M2 (PKM2) and prohibitin 2 (PHB2) interaction followed by MQC impairment in the pathogenesis of SC. We utilized LPS-induced SC models in PKM2 transgenic (PKM2TG) mice, PHB2S91D-knockin mice, and PKM2-overexpressing HL-1 cardiomyocytes. After LPS-induced SC, cardiac PKM2 expression was significantly downregulated in wild-type mice, whereas PKM2 overexpression in vivo sustained heart function, suppressed myocardial inflammation, and attenuated cardiomyocyte death. PKM2 overexpression relieved sepsis-related mitochondrial damage via MQC normalization, evidenced by balanced mitochondrial fission/fusion, activated mitophagy, restored mitochondrial biogenesis, and inhibited mitochondrial unfolded protein response. Docking simulations, co-IP, and domain deletion mutant protein transfection experiments showed that PKM2 phosphorylates PHB2 at Ser91, preventing LPS-mediated PHB2 degradation. Additionally, the A domain of PKM2 and the PHB domain of PHB2 are required for PKM2-PHB2 binding and PHB2 phosphorylation. After LPS exposure, expression of a phosphorylation-defective PHB2S91A mutant negated the protective effects of PKM2 overexpression. Moreover, knockin mice expressing a phosphorylation-mimetic PHB2S91D mutant showed improved heart function, reduced inflammation, and preserved mitochondrial function following sepsis induction. Abundant PKM2 expression is a prerequisite to sustain PKM2-PHB2 interaction which is a key element for preservation of PHB2 phosphorylation and MQC, presenting novel interventive targets for the treatment of septic cardiomyopathy.


Sujet(s)
Cardiomyopathies , Myocytes cardiaques , Prohibitines , Pyruvate kinase , Protéines de répression , Sepsie , Animaux , Phosphorylation , Cardiomyopathies/métabolisme , Cardiomyopathies/anatomopathologie , Souris , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique , Sepsie/métabolisme , Protéines de répression/métabolisme , Protéines de répression/génétique , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Mitochondries du myocarde/métabolisme , Souris transgéniques , Souris de lignée C57BL , Mâle , Lipopolysaccharides , Humains , Mitophagie
2.
FASEB J ; 38(11): e23702, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38837439

RÉSUMÉ

Pyruvate kinase is a glycolytic enzyme that converts phosphoenolpyruvate and ADP into pyruvate and ATP. There are two genes that encode pyruvate kinase in vertebrates; Pkm and Pkl encode muscle- and liver/erythrocyte-specific forms, respectively. Each gene encodes two isoenzymes due to alternative splicing. Both muscle-specific enzymes, PKM1 and PKM2, function in glycolysis, but PKM2 also has been implicated in gene regulation due to its ability to phosphorylate histone 3 threonine 11 (H3T11) in cancer cells. Here, we examined the roles of PKM1 and PKM2 during myoblast differentiation. RNA-seq analysis revealed that PKM2 promotes the expression of Dpf2/Baf45d and Baf250a/Arid1A. DPF2 and BAF250a are subunits that identify a specific sub-family of the mammalian SWI/SNF (mSWI/SNF) of chromatin remodeling enzymes that is required for the activation of myogenic gene expression during differentiation. PKM2 also mediated the incorporation of DPF2 and BAF250a into the regulatory sequences controlling myogenic gene expression. PKM1 did not affect expression but was required for nuclear localization of DPF2. Additionally, PKM2 was required not only for the incorporation of phosphorylated H3T11 in myogenic promoters but also for the incorporation of phosphorylated H3T6 and H3T45 at myogenic promoters via regulation of AKT and protein kinase C isoforms that phosphorylate those amino acids. Our results identify multiple unique roles for PKM2 and a novel function for PKM1 in gene expression and chromatin regulation during myoblast differentiation.


Sujet(s)
Différenciation cellulaire , Histone , Myoblastes , Pyruvate kinase , Animaux , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique , Souris , Phosphorylation , Histone/métabolisme , Histone/génétique , Myoblastes/métabolisme , Myoblastes/cytologie , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , , Humains , Protéines chromosomiques nonhistones/métabolisme , Protéines chromosomiques nonhistones/génétique , Hormones thyroïdiennes/métabolisme , Hormones thyroïdiennes/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Isoenzymes/métabolisme , Isoenzymes/génétique
3.
Int J Med Sci ; 21(6): 983-993, 2024.
Article de Anglais | MEDLINE | ID: mdl-38774750

RÉSUMÉ

Previous studies have highlighted the protective effects of pyruvate kinase M2 (PKM2) overexpression in septic cardiomyopathy. In our study, we utilized cardiomyocyte-specific PKM2 knockout mice to further investigate the role of PKM2 in attenuating LPS-induced myocardial dysfunction, focusing on mitochondrial biogenesis and prohibitin 2 (PHB2). Our findings confirmed that the deletion of PKM2 in cardiomyocytes significantly exacerbated LPS-induced myocardial dysfunction, as evidenced by impaired contractile function and relaxation. Additionally, the deletion of PKM2 intensified LPS-induced myocardial inflammation. At the molecular level, LPS triggered mitochondrial dysfunction, characterized by reduced ATP production, compromised mitochondrial respiratory complex I/III activities, and increased ROS production. Intriguingly, the absence of PKM2 further worsened LPS-induced mitochondrial damage. Our molecular investigations revealed that LPS disrupted mitochondrial biogenesis in cardiomyocytes, a disruption that was exacerbated by the absence of PKM2. Given that PHB2 is known as a downstream effector of PKM2, we employed PHB2 adenovirus to restore PHB2 levels. The overexpression of PHB2 normalized mitochondrial biogenesis, restored mitochondrial integrity, and promoted mitochondrial function. Overall, our results underscore the critical role of PKM2 in regulating the progression of septic cardiomyopathy. PKM2 deficiency impeded mitochondrial biogenesis, leading to compromised mitochondrial integrity, increased myocardial inflammation, and impaired cardiac function. The overexpression of PHB2 mitigated the deleterious effects of PKM2 deletion. This discovery offers a novel insight into the molecular mechanisms underlying septic cardiomyopathy and suggests potential therapeutic targets for intervention.


Sujet(s)
Cardiomyopathies , Souris knockout , Mitochondries du myocarde , Myocytes cardiaques , Prohibitines , Pyruvate kinase , Sepsie , Animaux , Cardiomyopathies/anatomopathologie , Cardiomyopathies/métabolisme , Cardiomyopathies/génétique , Cardiomyopathies/étiologie , Souris , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/métabolisme , Sepsie/métabolisme , Sepsie/anatomopathologie , Sepsie/génétique , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique , Mitochondries du myocarde/métabolisme , Mitochondries du myocarde/anatomopathologie , Protéines de répression/génétique , Protéines de répression/métabolisme , Humains , Biogenèse des organelles , Lipopolysaccharides/toxicité , Mâle , Modèles animaux de maladie humaine
4.
Fluids Barriers CNS ; 21(1): 42, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38755642

RÉSUMÉ

BACKGROUND: Most subarachnoid hemorrhage (SAH) patients have no obvious hematoma lesions but exhibit blood-brain barrier dysfunction and vasogenic brain edema. However, there is a few days between blood‒brain barrier dysfunction and vasogenic brain edema. The present study sought to investigate whether this phenomenon is caused by endothelial injury induced by the acute astrocytic barrier, also known as the glial limitans. METHODS: Bioinformatics analyses of human endothelial cells and astrocytes under hypoxia were performed based on the GEO database. Wild-type, EGLN3 and PKM2 conditional knock-in mice were used to confirm glial limitan formation after SAH. Then, the effect of endothelial EGLN3-PKM2 signaling on temporal and spatial changes in glial limitans was evaluated in both in vivo and in vitro models of SAH. RESULTS: The data indicate that in the acute phase after SAH, astrocytes can form a temporary protective barrier, the glia limitans, around blood vessels that helps maintain barrier function and improve neurological prognosis. Molecular docking studies have shown that endothelial cells and astrocytes can promote glial limitans-based protection against early brain injury through EGLN3/PKM2 signaling and further activation of the PKC/ERK/MAPK signaling pathway in astrocytes after SAH. CONCLUSION: Improving the ability to maintain glial limitans may be a new therapeutic strategy for improving the prognosis of SAH patients.


Sujet(s)
Astrocytes , Barrière hémato-encéphalique , Cellules endothéliales , Transduction du signal , Hémorragie meningée , Animaux , Astrocytes/métabolisme , Humains , Hémorragie meningée/métabolisme , Hémorragie meningée/immunologie , Souris , Transduction du signal/physiologie , Barrière hémato-encéphalique/métabolisme , Cellules endothéliales/métabolisme , Souris de lignée C57BL , Mâle , Pyruvate kinase/métabolisme , Protéines de transport/métabolisme , Oedème cérébral/métabolisme , Souris transgéniques , Protéines membranaires/métabolisme
5.
Plant Mol Biol ; 114(3): 60, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38758412

RÉSUMÉ

Pyruvate kinase (Pyk, EC 2.7.1.40) is a glycolytic enzyme that generates pyruvate and adenosine triphosphate (ATP) from phosphoenolpyruvate (PEP) and adenosine diphosphate (ADP), respectively. Pyk couples pyruvate and tricarboxylic acid metabolisms. Synechocystis sp. PCC 6803 possesses two pyk genes (encoded pyk1, sll0587 and pyk2, sll1275). A previous study suggested that pyk2 and not pyk1 is essential for cell viability; however, its biochemical analysis is yet to be performed. Herein, we biochemically analyzed Synechocystis Pyk2 (hereafter, SyPyk2). The optimum pH and temperature of SyPyk2 were 7.0 and 55 °C, respectively, and the Km values for PEP and ADP under optimal conditions were 1.5 and 0.053 mM, respectively. SyPyk2 is activated in the presence of glucose-6-phosphate (G6P) and ribose-5-phosphate (R5P); however, it remains unaltered in the presence of adenosine monophosphate (AMP) or fructose-1,6-bisphosphate. These results indicate that SyPyk2 is classified as PykA type rather than PykF, stimulated by sugar monophosphates, such as G6P and R5P, but not by AMP. SyPyk2, considering substrate affinity and effectors, can play pivotal roles in sugar catabolism under nonphotosynthetic conditions.


Sujet(s)
Glucose-6-phosphate , Phosphoénolpyruvate , Pyruvate kinase , Ribose monophosphate , Synechocystis , Synechocystis/métabolisme , Synechocystis/génétique , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique , Phosphoénolpyruvate/métabolisme , Glucose-6-phosphate/métabolisme , Ribose monophosphate/métabolisme , Spécificité du substrat , Concentration en ions d'hydrogène , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Cinétique , Température
6.
Bioorg Chem ; 147: 107425, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38714117

RÉSUMÉ

Non-alcoholic fatty liver disease (NAFLD) comprises a broad range of liver disease including hepatocellular carcinoma (HCC) with is no FDA-approved drug. Liver pyruvate kinase (PKL) is a major regulator of metabolic flux and ATP generation in liver presenting a potential target for the treatment of NAFLD. Based on our recent finding of JNK-5A's effectiveness in inhibiting PKLR expression through a drug repositioning pipeline, this study aims to improve its efficacy further. We synthesized a series of JNK-5A analogues with targeted modifications, guided by molecular docking studies. These compounds were evaluated for their activities on PKL expression, cell viability, triacylglyceride (TAG) levels, and the expressions of steatosis-related proteins in the human HepG2 cell line. Subsequently, the efficacy of these compounds was assessed in reducing TAG level and toxicity. Compounds 40 (SET-151) and 41 (SET-152) proved to be the most efficient in reducing TAG levels (11.51 ± 0.90 % and 10.77 ± 0.67 %) and demonstrated lower toxicity (61.60 ± 5.00 % and 43.87 ± 1.42 %) in HepG2 cells. Additionally, all synthesized compounds were evaluated for their anti-cancer properties revealing that compound 74 (SET-171) exhibited the highest toxicity in cell viability with IC50 values of 8.82 µM and 2.97 µM in HepG2 and Huh7 cell lines, respectively. To summarize, compounds 40 (SET-151) and 41 (SET-152) show potential for treating NAFLD, while compound 74 (SET-171) holds potential for HCC therapy.


Sujet(s)
Carcinome hépatocellulaire , Conception de médicament , Tumeurs du foie , Stéatose hépatique non alcoolique , Inhibiteurs de protéines kinases , Humains , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/anatomopathologie , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Stéatose hépatique non alcoolique/traitement médicamenteux , Relation structure-activité , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/composition chimique , Cellules HepG2 , Structure moléculaire , Pyruvate kinase/antagonistes et inhibiteurs , Pyruvate kinase/métabolisme , Simulation de docking moléculaire , Relation dose-effet des médicaments , Survie cellulaire/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique
7.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 636-643, 2024 Apr 20.
Article de Chinois | MEDLINE | ID: mdl-38708495

RÉSUMÉ

OBJECTIVE: To investigate the effect of Jisuikang formula-medicated serum for promoting spinal cord injury (SCI) repair in rats and explore the possible mechanism. METHODS: Thirty adult SD rats were randomized into sham-operated group, SCI (induced using a modified Allen method) model group, and Jisuikang formula-medicated serum treatment group. After the operations, the rats were treated with normal saline or Jisuikang by gavage on a daily basis for 14 days, and the changes in hindlimb motor function of the rats was assessed with Basso-Beattie-Bresnahan (BBB) scores and inclined-plate test. The injured spinal cord tissues were sampled from the SCI rat models for single-cell RNA sequencing, and bioinformatics analysis was performed to identify the target genes of Jisuikang, spinal cord injury and glycolysis. In the cell experiment, cultured astrocytes from neonatal SD rat cortex were treated with SOX2 alone or in combination with Jisuikang-medicated serum for 21 days, and the protein expressions of PKM2, p-PKM2 and YAP and colocalization of PKM2 and YAP in the cells were analyzed with Western blotting and immunofluorescence staining, respectively. RESULTS: The SCI rats with Jisuikang treatment showed significantly improved BBB scores and performance in inclined-plate test. At the injury site, high PKM2 expression was detected in various cell types. Bioinformatic analysis identified the HIPPO-YAP signaling pathway as the target pathway of Jisuikang. In cultured astrocytes, SOX2 combined with the mediated serum, as compared with SOX2 alone, significantly increased PKM2, p-PKM2 and YAP expressions and entry of phosphorylated PKM2 into the nucleus, and promoted PKM2 and YAP co-localization in the cells. CONCLUSION: Jisuikang formula accelerates SCI repair in rats possibly by promoting aerobic glycolysis of the astrocytes via activating the PKM2/YAP axis to induce reprogramming of the astrocytes into neurons.


Sujet(s)
Astrocytes , Pyruvate kinase , Transduction du signal , Traumatismes de la moelle épinière , Protéines de signalisation YAP , Animaux , Rats , Astrocytes/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Protéines de transport/métabolisme , Modèles animaux de maladie humaine , Médicaments issus de plantes chinoises/pharmacologie , Protéines membranaires/métabolisme , Rat Sprague-Dawley , Transduction du signal/effets des médicaments et des substances chimiques , Traumatismes de la moelle épinière/métabolisme , Traumatismes de la moelle épinière/traitement médicamenteux , , Hormones thyroïdiennes/métabolisme
8.
Cells ; 13(10)2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38786060

RÉSUMÉ

Cells defend against oxidative stress by enhancing antioxidant capacity, including stress-activated metabolic alterations, but the underlying intracellular signaling mechanisms remain unclear. This paper reports that immunoglobulin superfamily containing leucine-rich repeat (ISLR) functions as a redox sensor that responds to reactive oxygen species (ROS) stimulation and modulates the antioxidant capacity by suppressing pyruvate kinase isozyme M2 (PKM2) activity. Following oxidative stress, ISLR perceives ROS stimulation through its cysteine residue 19, and rapidly degrades in the autophagy-lysosome pathway. The downregulated ISLR enhances the antioxidant capacity by promoting the tetramerization of PKM2, and then enhancing the pyruvate kinase activity, PKM2-mediated glycolysis is crucial to the ISLR-mediated antioxidant capacity. In addition, our results demonstrated that, in triple-negative breast cancer, cisplatin treatment reduced the level of ISLR, and PKM2 inhibition sensitizes tumors to cisplatin by enhancing ROS production; and argued that PKM2 inhibition can synergize with cisplatin to limit tumor growth. Our results demonstrate a molecular mechanism by which cells respond to oxidative stress and modulate the redox balance.


Sujet(s)
Antioxydants , Oxydoréduction , Stress oxydatif , Espèces réactives de l'oxygène , Humains , Oxydoréduction/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Lignée cellulaire tumorale , Antioxydants/métabolisme , Antioxydants/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Animaux , Cisplatine/pharmacologie , Femelle , Protéines membranaires/métabolisme , Hormones thyroïdiennes/métabolisme , , Souris , Pyruvate kinase/métabolisme , Glycolyse/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Protéines de transport/métabolisme , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/enzymologie
10.
Biomolecules ; 14(5)2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38785973

RÉSUMÉ

One of the hallmarks of cancer is metabolic reprogramming in tumor cells, and aerobic glycolysis is the primary mechanism by which glucose is quickly transformed into lactate. As one of the primary rate-limiting enzymes, pyruvate kinase (PK) M is engaged in the last phase of aerobic glycolysis. Alternative splicing is a crucial mechanism for protein diversity, and it promotes PKM precursor mRNA splicing to produce PKM2 dominance, resulting in low PKM1 expression. Specific splicing isoforms are produced in various tissues or illness situations, and the post-translational modifications are linked to numerous disorders, including cancers. hnRNPs are one of the main components of the splicing factor families. However, there have been no comprehensive studies on hnRNPs regulating PKM alternative splicing. Therefore, this review focuses on the regulatory network of hnRNPs on PKM pre-mRNA alternative splicing in tumors and clinical drug research. We elucidate the role of alternative splicing in tumor progression, prognosis, and the potential mechanism of abnormal RNA splicing. We also summarize the drug targets retarding tumorous splicing events, which may be critical to improving the specificity and effectiveness of current therapeutic interventions.


Sujet(s)
Épissage alternatif , Ribonucléoprotéines nucléaires hétérogènes , Tumeurs , Pyruvate kinase , Humains , Épissage alternatif/génétique , Tumeurs/génétique , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Pyruvate kinase/génétique , Pyruvate kinase/métabolisme , Évolution de la maladie , Régulation de l'expression des gènes tumoraux , Animaux
11.
Neurol Res ; 46(7): 583-592, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38797679

RÉSUMÉ

BACKGROUND: Glioma is a common intracranial tumor, exhibiting a high degree of aggressiveness and invasiveness. Pyruvate kinase M2 (PKM2) is overexpressed in glioma tissues. However, the biological role of PKM2 in glioma is unclear. METHODS: The qRT-PCR, CCK-8, Transwell, flow cytometry detection, western blot assays, ELISA assay, and pyruvate kinase activity assays were performed in glioma cells transfected with PKM2 shRNA to explore the function of PKM2 in glioma progression. Then, STRING website was used to predict the proteins that interacted with PKM2, and Co-IP assay was conducted to further validate their interaction. Subsequently, the above experiments were performed again to find the effect of catenin beta 1 (CTNNB1) overexpression on PKM2-deficient glioma cells. The transplanted tumor models were also established to further validate our findings. RESULTS: PKM2 was up-regulated in glioma cells and tissues. After inhibiting PKM2, the proliferation, migration, glycolysis, and EMT of glioma cells were significantly decreased, and the proportion of apoptosis was increased. The prediction results of STRING website showed that CTNNB1 and PKM2 had the highest interaction score. The correlation between CTNNB1 and PKM2 was further confirmed by Co-IP test. PKM2 knockdown suppressed glioma cell proliferation, migration, glycolysis, and EMT, while CTNNB1 overexpression rescued these inhibitory effects. Correspondingly, PKM2 knockdown inhibited glioma growth in vivo. CONCLUSION: In summary, these findings indicated that PKM2 promotes glioma progression by mediating CTNNB1 expression, providing a possible molecular marker for the clinical management of gliomas.


Sujet(s)
Tumeurs du cerveau , Prolifération cellulaire , Évolution de la maladie , Gliome , , Hormones thyroïdiennes , bêta-Caténine , Gliome/anatomopathologie , Gliome/génétique , Gliome/métabolisme , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Humains , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Lignée cellulaire tumorale , Animaux , Hormones thyroïdiennes/métabolisme , Hormones thyroïdiennes/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Souris , Protéines de transport/métabolisme , Protéines de transport/génétique , Souris nude , Mouvement cellulaire/physiologie , Apoptose/physiologie , Régulation de l'expression des gènes tumoraux , Mâle , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique
12.
Cell Rep Methods ; 4(5): 100764, 2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38714198

RÉSUMÉ

Co-assembling enzymes with nanoparticles (NPs) into nanoclusters allows them to access channeling, a highly efficient form of multienzyme catalysis. Using pyruvate kinase (PykA) and lactate dehydrogenase (LDH) to convert phosphoenolpyruvic acid to lactic acid with semiconductor quantum dots (QDs) confirms how enzyme cluster formation dictates the rate of coupled catalytic flux (kflux) across a series of differentially sized/shaped QDs and 2D nanoplatelets (NPLs). Enzyme kinetics and coupled flux were used to demonstrate that by mixing different NP systems into clusters, a >10× improvement in kflux is observed relative to free enzymes, which is also ≥2× greater than enhancement on individual NPs. Cluster formation was characterized with gel electrophoresis and transmission electron microscopy (TEM) imaging. The generalizability of this mixed-NP approach to improving flux is confirmed by application to a seven-enzyme system. This represents a powerful approach for accessing channeling with almost any choice of enzymes constituting a multienzyme cascade.


Sujet(s)
L-Lactate dehydrogenase , Acide lactique , Nanoparticules , Phosphoénolpyruvate , Pyruvate kinase , L-Lactate dehydrogenase/métabolisme , L-Lactate dehydrogenase/composition chimique , Acide lactique/métabolisme , Acide lactique/composition chimique , Pyruvate kinase/métabolisme , Pyruvate kinase/composition chimique , Nanoparticules/composition chimique , Phosphoénolpyruvate/métabolisme , Boîtes quantiques/composition chimique , Cinétique
13.
Eur J Med Chem ; 272: 116426, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38718622

RÉSUMÉ

Pyruvate kinase isoform 2 (PKM2) is closely related to the regulation of Th17/Treg balance, which is considered to be an effective strategy for UC therapy. Parthenolide (PTL), a natural product, only possesses moderate PKM2-activating activity. Thus, five series of PTL derivatives are designed and synthesized to improve PKM2-activated activities and anti-UC abilities. Through detailed structure optimization, B4 demonstrates potent T-cell anti-proliferation activity (IC50 = 0.43 µM) and excellent PKM2-activated ability (AC50 = 0.144 µM). Subsequently, through mass spectrometry analysis, B4 is identified to interact with Cys423 of PKM2 via covalent-bond. Molecular docking and molecular dynamic simulation results reveal that the trifluoromethoxy of B4 forms a stronger hydrophobic interaction with Ala401, Pro402, and Ile403. In addition, B4 has a significant effect only on Th17 cell differentiation, thereby regulating the Th17/Treg balance. The effect of B4 on Th17/Treg imbalance can be attributed to inhibition of PKM2 dimer translocation and suppression of glucose metabolism. Finally, B4 can notably ameliorate the symptoms of dextran sulfate sodium (DSS)-induced colitis in mouse model in vivo. Thus, B4 is confirmed as a potent PKM2 activator, and has the potential to develop as a novel anti-UC agent.


Sujet(s)
Rectocolite hémorragique , Conception de médicament , Lactones , Pyruvate kinase , Sesquiterpènes , Sesquiterpènes/pharmacologie , Sesquiterpènes/composition chimique , Sesquiterpènes/synthèse chimique , Animaux , Souris , Pyruvate kinase/métabolisme , Pyruvate kinase/antagonistes et inhibiteurs , Lactones/pharmacologie , Lactones/composition chimique , Lactones/synthèse chimique , Relation structure-activité , Rectocolite hémorragique/traitement médicamenteux , Rectocolite hémorragique/induit chimiquement , Humains , Structure moléculaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Relation dose-effet des médicaments , Mâle , Sulfate dextran , Simulation de docking moléculaire , Hormones thyroïdiennes/métabolisme , Cellules Th17/effets des médicaments et des substances chimiques ,
14.
J Biosci Bioeng ; 138(1): 29-35, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38719683

RÉSUMÉ

Recombinase polymerase amplification (RPA) is an isothermal DNA amplification reaction at around 41 °C using recombinase (Rec), single-stranded DNA-binding protein (SSB), strand-displacing DNA polymerase (Pol), and an ATP-regenerating enzyme. Considering the onsite use of RPA reagents, lyophilized RPA reagents with long storage stability are highly desired. In this study, as one of the approaches to solve this problem, we attempted to use a thermostable pyruvate kinase (PK). PK gene was isolated from a thermophilic bacterium Thermotoga maritima (Tma-PK). Tma-PK was expressed in Escherichia coli and purified from the cells. Tma-PK exhibited higher thermostability than human PK. The purified Tma-PK preparation was applied to RPA as an ATP-regenerating enzyme. Liquid RPA reagent with Tma-PK exhibited the same performance as that with human PK. Lyophilized RPA reagent with Tma-PK exhibited higher performance than that with human PK. Combined with our previous results of RPA reagents of thermostable Pol from a thermophilic bacterium, Aeribacillus pallidus, the results in this study suggest that thermostable enzymes are preferable to mesophilic ones as a component in lyophilized RPA reagents.


Sujet(s)
Stabilité enzymatique , Lyophilisation , Techniques d'amplification d'acides nucléiques , Pyruvate kinase , Thermotoga maritima , Thermotoga maritima/enzymologie , Thermotoga maritima/génétique , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique , Pyruvate kinase/composition chimique , Techniques d'amplification d'acides nucléiques/méthodes , Humains , Recombinases/métabolisme , Recombinases/composition chimique , Recombinases/génétique , Escherichia coli/génétique , Escherichia coli/métabolisme , DNA-directed DNA polymerase/métabolisme , DNA-directed DNA polymerase/composition chimique , DNA-directed DNA polymerase/génétique , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Protéines bactériennes/composition chimique
15.
Free Radic Biol Med ; 221: 52-63, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38754745

RÉSUMÉ

OBJECTIVE: Virus infection is a major threat to human health and remains a significant cause of death to date. Macrophages are important innate immune cells that exhibit indispensable roles in controlling virus replication. It was recently reported that metabolic adaption determines the functional state of macrophages. Thus, to further unravel the crucial factors involving in metabolic adaption of macrophages might provide the potential candidates for optimizing their anti-viral capabilities. METHODS: RT-PCR, Western blotting, virus plaque assay and HE were used to evaluate the viral load in virus-infected Tipe1M-KO and Tipe1f/f mice or cultured macrophages. RNA sequencing were performed with Tipe1M-KOor Tipe1f/f BMDMs upon virus infection. Extracellular acidification rate (ECAR) was applied for analyzing glycolysis rate in virus-infected BMDMs. Co-immunoprecipitation (Co-IP) assay and LC-MS/MS were used to determine the potential interacting proteins of TIPE1. RESULTS: TIPE1 level was significantly reduced in BMDMs infected with either RNA viruses or DNA virus. Deficiency of Tipe1 in macrophages increased viral load and aggravated tissue damage. Mechanistically, TIPE1 suppressed the glycolytic capacity of macrophages through interacting with PKM2 and promoting its ubiquitination degradation, which in turn decreased HIF1α transcription and viral replication in macrophages. CONCLUSIONS: TIPE1 functions as a novel regulator for metabolic reprogramming and virus infection in macrophages.


Sujet(s)
Glycolyse , Sous-unité alpha du facteur-1 induit par l'hypoxie , Protéines et peptides de signalisation intracellulaire , Macrophages , Protéines membranaires , , Réplication virale , Animaux , Souris , Glycolyse/génétique , Réplication virale/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Macrophages/virologie , Macrophages/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Protéines de transport/métabolisme , Protéines de transport/génétique , Humains , Souris knockout , Ubiquitination , Rétrocontrôle physiologique , Souris de lignée C57BL , Pyruvate kinase
16.
J Agric Food Chem ; 72(20): 11724-11732, 2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38718268

RÉSUMÉ

Protein post-translational modifications (PTMs) play an essential role in meat quality development. However, the effect of specific PTM sites on meat proteins has not been investigated yet. The characteristics of pyruvate kinase M (PKM) were found to exhibit a close correlation with final meat quality, and thus, serine 99 (S99) and lysine 137 (K137) in PKM were mutated to study their effect on PKM function. The structural and functional properties of five lamb PKM variants, including wild-type PKM (wtPKM), PKM_S99D (S99 phosphorylation), PKM_S99A (PKM S99 dephosphorylation), PKM_K137Q (PKM K137 acetylation), and PKM_K137R (PKM K137 deacetylation), were evaluated. The results showed that the secondary structure, tertiary structure, and polymer formation were affected among different PKM variants. In addition, the glycolytic activity of PKM_K137Q was decreased because of its weakened binding with phosphoenolpyruvate. In the PKM_K137R variant, the actin phosphorylation level exhibited a decrease, suggesting a low kinase activity of PKM_K137R. The results of molecular simulation showed a 42% reduction in the interface area between PKM_K137R and actin, in contrast to wtPKM and actin. These findings are significant for revealing the mechanism of how PTMs regulate PKM function and provide a theoretical foundation for the development of precise meat quality preservation technology.


Sujet(s)
Glycolyse , Pyruvate kinase , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique , Pyruvate kinase/composition chimique , Phosphorylation , Animaux , Acétylation , Ovis , Maturation post-traductionnelle des protéines , Protein kinases/métabolisme , Protein kinases/génétique , Protein kinases/composition chimique , Viande/analyse
17.
Biochem Biophys Res Commun ; 721: 150130, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38761750

RÉSUMÉ

Apigenin (API) is a natural flavonoid compound with antioxidant, anti fibrotic, anti-inflammatory and other effects, but there is limited research on the effect of API on liver fibrosis. This study aims to explore the effect and potential mechanism of API on liver fibrosis induced by CCl4 in mice. The results indicate that API reduces oxidative stress levels, inhibits hepatic stellate cell (HSC) activation, and exerts anti liver fibrosis effects by regulating the PKM2-HIF-1α pathway. We observed that API alleviated liver tissue pathological damage and collagen deposition in CCl4 induced mouse liver fibrosis model, promoting the recovery of liver function in mice with liver fibrosis. In addition, the API inhibits the transition of Pyruvate kinase isozyme type M2 (PKM2) from dimer to tetramer formation by regulating the EGFR-MEK1/2-ERK1/2 pathway, thereby preventing dimer from entering the nucleus and blocking PKM2-HIF-1α access. This change leads to a decrease in malondialdehyde (MDA) and Catalase (CAT) levels and an increase in glutathione (GSH), superoxide dismutase (SOD), glutathione peroxidase (GSH-PX) levels, as well as total antioxidant capacity (T-AOC) in the liver of liver fibrosis mice. At the same time, API downregulated the expression of α-smooth muscle actin (α-SMA), Vimentin and Desmin in the liver tissue of mice with liver fibrosis, inhibited the activation of HSC, and reduced collagen deposition. These results indicate that API can inhibit HSC activation and alleviate CCl4 induced liver fibrosis by inhibiting the PKM2-HIF-1α pathway and reducing oxidative stress, laying an important foundation for the development and clinical application of API as a novel drug for treating liver fibrosis.


Sujet(s)
Apigénine , Sous-unité alpha du facteur-1 induit par l'hypoxie , Cirrhose du foie , Stress oxydatif , Animaux , Stress oxydatif/effets des médicaments et des substances chimiques , Apigénine/pharmacologie , Apigénine/usage thérapeutique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Cirrhose du foie/métabolisme , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/anatomopathologie , Souris , Mâle , Pyruvate kinase/métabolisme , Souris de lignée C57BL , Tétrachloro-méthane/toxicité , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Cellules étoilées du foie/anatomopathologie , , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Hormones thyroïdiennes/métabolisme , Antioxydants/pharmacologie , Antioxydants/métabolisme , Récepteurs ErbB
18.
BMC Cancer ; 24(1): 511, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38654205

RÉSUMÉ

BACKGROUND: Although papillary thyroid carcinoma (PTC) has a favorable prognosis, it could affect patient life quality and become a serious threat because of invasion and metastasis. Many investigations have suggested that circular RNAs (circRNAs) are involved in different cancer regulations. Nevertheless, circRNAs role in invasive PTC remains unclear. METHODS: In the present investigation, next-generation sequencing was applied to explore abnormal circRNA expression. The expression of circRNA phosphoglycerate dehydrogenase (circPHGDH) in PTC cell lines and tissues were examined. Then, we investigated regulatory mechanism and circPHGDH downstream targets using bioinformatics analysis and luciferase reporting analysis. Then transwell migration, Cell Counting Kit-8 (CCK8) and 5-ethynyl-2'-deoxyuridine (EdU) assays were used for cells migration and proliferation analysis. In vivo metastasis and tumorigenesis assays were also employed to evaluate the circPHGDH role in PTC. RESULTS: The data showcased that circPHGDH expression increased in both PTC cell lines and tissues, which suggested that circPHGDH functions in PTC progression. circPHGDH downregulation suppressed PTC invasion and proliferation in both in vivo and in vitro experiments. Bioinformatics and luciferase reporter results confirmed that both microRNA (miR)-122-5p and pyruvate kinase M2 subtype (PKM2) were downstream targets of circPHGDH. PKM2 overexpression or miR-122-5p suppression reversed PTC cell invasion and proliferation post silencing circPHGDH by restoring aerobic glycolysis. CONCLUSION: Taken together, our research found that circPHGDH downregulation reduced PTC progression via miR-122-5p/PKM2 axis regulation mediated by aerobic glycolysis.


Sujet(s)
Prolifération cellulaire , Évolution de la maladie , Régulation négative , Protéines membranaires , microARN , Phosphoglycerate dehydrogenase , ARN circulaire , Cancer papillaire de la thyroïde , Tumeurs de la thyroïde , Animaux , Femelle , Humains , Mâle , Souris , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Souris nude , microARN/génétique , microARN/métabolisme , Invasion tumorale , Phosphoglycerate dehydrogenase/génétique , ARN circulaire/génétique , ARN circulaire/métabolisme , Cancer papillaire de la thyroïde/génétique , Cancer papillaire de la thyroïde/anatomopathologie , Cancer papillaire de la thyroïde/métabolisme , Tumeurs de la thyroïde/génétique , Tumeurs de la thyroïde/anatomopathologie , Tumeurs de la thyroïde/métabolisme , Pyruvate kinase/génétique , Pyruvate kinase/métabolisme
19.
Front Immunol ; 15: 1371706, 2024.
Article de Anglais | MEDLINE | ID: mdl-38650935

RÉSUMÉ

Pulmonary hypertension (PH) pathogenesis is driven by inflammatory and metabolic derangements as well as glycolytic reprogramming. Induction of both interleukin 6 (IL6) and transglutaminase 2 (TG2) expression participates in human and experimental cardiovascular diseases. However, little is known about the role of TG2 in these pathologic processes. The current study aimed to investigate the molecular interactions between TG2 and IL6 in mediation of tissue remodeling in PH. A lung-specific IL6 over-expressing transgenic mouse strain showed elevated right ventricular (RV) systolic pressure as well as increased wet and dry tissue weights and tissue fibrosis in both lungs and RVs compared to age-matched wild-type littermates. In addition, IL6 over-expression induced the glycolytic and fibrogenic markers, hypoxia-inducible factor 1α, pyruvate kinase M2 (PKM2), and TG2. Consistent with these findings, IL6 induced the expression of both glycolytic and pro-fibrogenic markers in cultured lung fibroblasts. IL6 also induced TG2 activation and the accumulation of TG2 in the extracellular matrix. Pharmacologic inhibition of the glycolytic enzyme, PKM2 significantly attenuated IL6-induced TG2 activity and fibrogenesis. Thus, we conclude that IL6-induced TG2 activity and cardiopulmonary remodeling associated with tissue fibrosis are under regulatory control of the glycolytic enzyme, PKM2.


Sujet(s)
Fibroblastes , Protéines G , Hypertension pulmonaire , Interleukine-6 , Poumon , Souris transgéniques , Protein glutamine gamma glutamyltransferase-2 , Pyruvate kinase , Transglutaminases , Animaux , Humains , Souris , Modèles animaux de maladie humaine , Fibroblastes/métabolisme , Fibrose , Protéines G/métabolisme , Protéines G/génétique , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/anatomopathologie , Hypertension pulmonaire/étiologie , Interleukine-6/métabolisme , Poumon/anatomopathologie , Poumon/immunologie , Poumon/métabolisme , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/anatomopathologie , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique , Transglutaminases/métabolisme , Transglutaminases/génétique
20.
Eur J Pharmacol ; 972: 176551, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38570082

RÉSUMÉ

Fibroblast-like synoviocytes (FLS) play an important role in rheumatoid arthritis (RA)-related swelling and bone damage. Therefore, novel targets for RA therapy in FLS are urgently discovered for improving pathologic phenomenon, especially joint damage and dyskinesia. Here, we suggested that pyruvate kinase M2 (PKM2) in FLS represented a pharmacological target for RA treatment by antimalarial drug artemisinin (ART). We demonstrated that ART selectively inhibited human RA-FLS and rat collagen-induced arthritis (CIA)-FLS proliferation and migration without observed toxic effects. In particular, the identification of targets revealed that PKM2 played a crucial role as a primary regulator of the cell cycle, leading to the heightened proliferation of RA-FLS. ART exhibited a direct interaction with PKM2, resulting in an allosteric modulation that enhances the lactylation modification of PKM2. This interaction further promoted the binding of p300, ultimately preventing the nuclear translocation of PKM2 and inducing cell cycle arrest at the S phase. In vivo, ART obviously suppressed RA-mediated synovial hyperplasia, bone damage and inflammatory response to further improve motor behavior in CIA-rats. Taken together, these findings indicate that directing interventions towards PKM2 in FLS could offer a hopeful avenue for pharmaceutical treatments of RA through the regulation of cell cycle via PKM2 lactylation.


Sujet(s)
Polyarthrite rhumatoïde , Prolifération cellulaire , Cellules synoviales , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/métabolisme , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Humains , Rats , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Pyruvate kinase/métabolisme , , Mâle , Hormones thyroïdiennes/métabolisme , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Thérapie moléculaire ciblée , Protéines membranaires/métabolisme , Protéines de transport/métabolisme , Bibliothèques de petites molécules/pharmacologie , Bibliothèques de petites molécules/composition chimique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...