Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.166
Filter
1.
J Biochem Mol Toxicol ; 38(8): e23799, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39132768

ABSTRACT

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.


Subject(s)
Colorectal Neoplasms , Humans , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , Thyroid Hormones/metabolism , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Estrogens/pharmacology , Thyroid Hormone-Binding Proteins , Estradiol/pharmacology , Apoptosis/drug effects , Glucose/metabolism , Carrier Proteins/metabolism , Pyruvate Kinase/metabolism , Pyruvate Kinase/antagonists & inhibitors , Pyruvate Kinase/genetics , Glycolysis/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Female
2.
Br J Haematol ; 205(2): 613-623, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39118415

ABSTRACT

Pyruvate kinase (PK) deficiency, a rare, congenital haemolytic anaemia caused by mutations in the PKLR gene, is associated with many clinical manifestations, but the full disease burden has yet to be characterised. The Peak Registry (NCT03481738) is an observational, longitudinal registry of adult and paediatric patients with PK deficiency. Here, we described comorbidities and complications in these patients by age at most recent visit and PKLR genotype. As of 13 May 2022, 241 patients were included in the analysis. In total, 48.3% had undergone splenectomy and 50.5% had received chelation therapy. History of iron overload (before enrolment/during follow-up) was common (52.5%), even in never-transfused patients (20.7%). Neonatal complications and symptoms included jaundice, splenomegaly and hepatomegaly, with treatment interventions required in 41.5%. Among adults, osteopenia/osteoporosis occurred in 19.0% and pulmonary hypertension in 6.7%, with median onset ages of 37, 33 and 22 years, respectively. Biliary events and bone health problems were common across PKLR genotypes. Among 11 patients who had thromboembolic events, eight had undergone prior splenectomy. Patients with PK deficiency may have many complications, which can occur early in and throughout life. Awareness of their high disease burden may help clinicians better provide appropriate monitoring and management of these patients.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic , Pyruvate Kinase , Pyruvate Metabolism, Inborn Errors , Registries , Humans , Pyruvate Kinase/deficiency , Pyruvate Kinase/genetics , Male , Female , Adult , Child , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Anemia, Hemolytic, Congenital Nonspherocytic/epidemiology , Pyruvate Metabolism, Inborn Errors/genetics , Pyruvate Metabolism, Inborn Errors/epidemiology , Adolescent , Child, Preschool , Infant , Comorbidity , Middle Aged , Splenectomy , Young Adult , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/epidemiology , Iron Overload/etiology , Iron Overload/epidemiology , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/epidemiology , Infant, Newborn
3.
Int J Chron Obstruct Pulmon Dis ; 19: 1905-1920, 2024.
Article in English | MEDLINE | ID: mdl-39206144

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Drugs, Chinese Herbal , Lung , Mitochondria , NF-E2-Related Factor 2 , Oxidative Stress , Pulmonary Disease, Chronic Obstructive , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/physiopathology , Humans , Oxidative Stress/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Lung/drug effects , Lung/metabolism , Lung/physiopathology , Lung/enzymology , Drugs, Chinese Herbal/pharmacology , A549 Cells , Male , Thyroid Hormone-Binding Proteins , Pyruvate Kinase/metabolism , Antioxidants/pharmacology , Rats, Sprague-Dawley , Carrier Proteins/metabolism , Thyroid Hormones/metabolism , Smoke/adverse effects , Membrane Proteins/metabolism , Membrane Proteins/genetics
4.
Mol Med Rep ; 30(4)2024 Oct.
Article in English | MEDLINE | ID: mdl-39155878

ABSTRACT

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.


Subject(s)
Pyruvate Kinase , Sepsis , Humans , Sepsis/metabolism , Pyruvate Kinase/metabolism , Glycolysis , Animals , Thyroid Hormone-Binding Proteins , Thyroid Hormones/metabolism
5.
Nat Commun ; 15(1): 7463, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39198443

ABSTRACT

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.


Subject(s)
Glucose , Hydrogen Sulfide , Hydrogen Sulfide/metabolism , Humans , Glucose/metabolism , Animals , Cell Line, Tumor , Mice , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics , Pyruvate Kinase/chemistry , Cysteine/metabolism , Glycolysis , Thyroid Hormones/metabolism , Mutation , Mitochondria/metabolism , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , Protein Multimerization , Mice, Nude , Carrier Proteins/metabolism , Carrier Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Thyroid Hormone-Binding Proteins
6.
J Agric Food Chem ; 72(32): 18013-18026, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39088205

ABSTRACT

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.


Subject(s)
Insulin Resistance , Lipid Metabolism , Muscle, Skeletal , Pyruvate Kinase , Xanthophylls , Animals , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Xanthophylls/pharmacology , Lipid Metabolism/drug effects , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics , Male , Humans , Mice, Inbred C57BL , Diet, High-Fat/adverse effects
7.
Cell Rep Med ; 5(8): 101684, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39128469

ABSTRACT

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.


Subject(s)
Brain , Homeostasis , Lactic Acid , Pyruvate Kinase , Sirtuin 1 , Sirtuin 1/metabolism , Sirtuin 1/genetics , Animals , Brain/metabolism , Brain/pathology , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics , Mice , Lactic Acid/metabolism , Humans , Acetylation/drug effects , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/genetics , Disease Models, Animal , Male , Mice, Inbred C57BL , Thyroid Hormone-Binding Proteins , Thyroid Hormones/metabolism , Naphthoquinones/pharmacology
8.
J Neuroimmune Pharmacol ; 19(1): 48, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39196455

ABSTRACT

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.


Subject(s)
Brain Injuries, Traumatic , Microglia , Mitochondria , Naphthoquinones , Pyruvate Kinase , Animals , Microglia/drug effects , Microglia/metabolism , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/drug therapy , Mice , Pyruvate Kinase/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Naphthoquinones/pharmacology , Male , Mice, Inbred C57BL , Protein Multimerization/drug effects , Cells, Cultured
9.
J Ethnopharmacol ; 335: 118679, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39121930

ABSTRACT

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.


Subject(s)
Fibroblasts , Fibrosis , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit , Kidney , Rats, Sprague-Dawley , Renal Insufficiency, Chronic , Thyroid Hormone-Binding Proteins , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Fibrosis/drug therapy , Fibroblasts/drug effects , Fibroblasts/metabolism , Male , Glycolysis/drug effects , Rats , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Drugs, Chinese Herbal/pharmacology , Cell Line , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Thyroid Hormones , Administration, Oral , Cell Proliferation/drug effects , Signal Transduction/drug effects , Carrier Proteins/metabolism , Carrier Proteins/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics
10.
Georgian Med News ; (349): 154-160, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38963220

ABSTRACT

Mitochondrial dysfunction in autism leads to impair the mitochondria's ability to synthesis adenosine triphosphate (ATP) by impairment citric acid cycle as well as increase anaerobic glycolysis. Aim - measuring and evaluating the levels of mitochondrial markers; including glutamate oxaloacetate transaminase (GOT), glutamate pyruvate transaminase (GPT), malate dehydrogenase, and pyruvate kinase) in the autistic group and knowing the possibility of using these markers to diagnose children with autism spectrum disorder. A case-control study was done in the Al-Zahraa Teaching Hospital (Kut City, Iraq) on 100 Iraqi children (male and female), between (April 2023 and January 2024). Their ages ranged between 3 and 9 years. Among them were 50 patients enrolled as autistic group and 50 healthy enrolled as control group. Blood samples were collected and bioassays for GOT, GPT, pyruvate kinase, and malate dehydrogenase were measured by ELISA technique. The autistic group showed that the urine GOT, urine GPT, serum malate, and serum pyruvate levels in the ASD group was significantly higher (P<0.001) than the control group. The ROC analysis showed that urine GOT, urine GOT, serum malate and serum pyruvate had an accuracy level of (81%,71%,77%, and 80 %) and the area under the curve (AUC) was > 0.7 (0.8),0.7, 0.7(0.76), and 0.7(0.8) thus urine GOT, urine GPT, serum, malate, and serum pyruvate are a valid diagnostic marker. There was a significant difference in the mean urine and serum concentrations of mitochondrial markers (GOT, GPT, malate dehydrogenase, and pyruvate kinase) between autistic children and the control group due to mitochondrial dysfunction.


Subject(s)
Aspartate Aminotransferases , Autism Spectrum Disorder , Biomarkers , Malate Dehydrogenase , Mitochondria , Pyruvate Kinase , Humans , Child , Male , Female , Autism Spectrum Disorder/blood , Malate Dehydrogenase/blood , Child, Preschool , Case-Control Studies , Pyruvate Kinase/blood , Biomarkers/blood , Biomarkers/urine , Aspartate Aminotransferases/blood , Mitochondria/metabolism , Alanine Transaminase/blood , Pyruvic Acid/blood , Malates/blood , ROC Curve
11.
J Coll Physicians Surg Pak ; 34(7): 811-816, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38978246

ABSTRACT

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.


Subject(s)
Carrier Proteins , Leukemia, Myeloid, Acute , Membrane Proteins , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Case-Control Studies , China/epidemiology , Leukemia, Myeloid, Acute/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Prognosis , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Thyroid Hormones/blood , Thyroid Hormones/metabolism
12.
Int J Mol Sci ; 25(14)2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39063228

ABSTRACT

Metabolic dysfunction-associated fatty liver disease (MAFLD) presents a significant global health challenge, characterized by the accumulation of liver fat and impacting a considerable portion of the worldwide population. Despite its widespread occurrence, effective treatments for MAFLD are limited. The liver-specific isoform of pyruvate kinase (PKL) has been identified as a promising target for developing MAFLD therapies. Urolithin C, an allosteric inhibitor of PKL, has shown potential in preliminary studies. Expanding upon this groundwork, our study delved into delineating the structure-activity relationship of urolithin C via the synthesis of sulfone-based urolithin analogs. Our results highlight that incorporating a sulfone moiety leads to substantial PKL inhibition, with additional catechol moieties further enhancing this effect. Despite modest improvements in liver cell lines, there was a significant increase in inhibition observed in HepG2 cell lysates. Specifically, compounds 15d, 9d, 15e, 18a, 12d, and 15a displayed promising IC50 values ranging from 4.3 µM to 18.7 µM. Notably, compound 15e not only demonstrated a decrease in PKL activity and triacylglycerol (TAG) content but also showed efficient cellular uptake. These findings position compound 15e as a promising candidate for pharmacological MAFLD treatment, warranting further research and studies.


Subject(s)
Liver , Pyruvate Kinase , Sulfones , Humans , Pyruvate Kinase/antagonists & inhibitors , Pyruvate Kinase/metabolism , Sulfones/chemistry , Sulfones/pharmacology , Sulfones/chemical synthesis , Hep G2 Cells , Liver/metabolism , Structure-Activity Relationship , Allosteric Regulation/drug effects , Drug Design , Coumarins/chemistry , Coumarins/pharmacology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis
13.
Int Immunopharmacol ; 139: 112675, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39024754

ABSTRACT

Pyruvate kinase M2 (PKM2), a key enzyme involved in glycolysis,plays an important role in regulating cell metabolism and growth under different physiological conditions. PKM2 has been intensively investigated in multiple cancer diseases. Recent years, many studies have found its pivotal role in cerebrovascular diseases (CeVDs), the disturbances in intracranial blood circulation. CeVDs has been confirmed to be closely associated with oxidative stress (OS), mitochondrial dynamics, systemic inflammation, and local neuroinflammation in the brain. It has further been revealed that PKM2 exerts various biological functions in the regulation of energy supply, OS, inflammatory responses, and mitochondrial dysfunction. The roles of PKM2 are closely related to its different isoforms, expression levels in subcellular localization, and post-translational modifications. Therefore, summarizing the roles of PKM2 in CeVDs will help further understanding the molecular mechanisms of CeVDs. In this review, we illustrate the characteristics of PKM2, the regulated PKM2 expression, and the biological roles of PKM2 in CeVDs.


Subject(s)
Cerebrovascular Disorders , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Humans , Animals , Cerebrovascular Disorders/metabolism , Thyroid Hormones/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Carrier Proteins/metabolism , Oxidative Stress , Pyruvate Kinase/metabolism , Brain/metabolism
14.
Technol Cancer Res Treat ; 23: 15330338241265983, 2024.
Article in English | MEDLINE | ID: mdl-39043046

ABSTRACT

Objective: Finding biomarkers related to non-small cell lung cancer (NSCLC) is helpful for the diagnosis and precise treatment of lung cancer. The relationship between serum tumor M2-pyruvate kinase (TuM2-PK), carcinoembryonic antigen (CEA), and cytokeratin 19 fragment (CYFRA21-1) and NSCLC was analyzed. Methods: The serum levels of TuM2-PK, CEA, and CYFRA21-1 in 184 patients with the NSCLC group, 60 patients with the benign lung disease (BLD) group, and 90 healthy controls (HC) group were detected. The levels of TuM2-PK were measured by using an enzyme-linked immunosorbent assay. The detection methods of CEA and CYFRA21-1 were electrochemiluminescence. The receiver operating characteristic (ROC) curve was drawn to evaluate the diagnostic value of TuM2-PK, CEA, and CYFRA21-1 on NSCLC. The Kaplan-Meier survival curve was drawn to evaluate the survival status in NSCLC patients with different serum levels of TuM2-PK, CEA, and CYFRA21-1. Results: Serum levels of TuM2-PK, CEA, and CYFRA21-1 in the NSCLC group were significantly higher than those in the BLD group and the HC group (P < .01). Serum levels of TuM2-PK, CEA, and CYFRA21-1 in NSCLC patients were associated with the tumor lymph node metastasis stage (P < .05), lymph node metastasis (P < .05), and distant metastasis (P < .05). The ROC curve showed that the area under the curve of serum levels of TuM2-PK, CEA, and CYFRA21-1 was 0.814, 0.638, and 0.719, respectively, and that the combination of the above 3 was 0.918. The Kaplan-Meier survival curve showed that the 1-, 3- and 5-year survival rate in NSCLC patients with positive TuM2-PK, CEA, and CYFRA21-1 was significantly lower than that in NSCLC patients with negative TuM2-PK, CEA, and CYFRA21-1, respectively (P < .05). Conclusions: Serum TuM2-PK, CEA, and CYFRA21-1 levels have high clinical values in the diagnosis of NSCLC, and can effectively judge the prognosis of patients.


Subject(s)
Antigens, Neoplasm , Biomarkers, Tumor , Carcinoembryonic Antigen , Carcinoma, Non-Small-Cell Lung , Keratin-19 , Lung Neoplasms , Pyruvate Kinase , ROC Curve , Humans , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/pathology , Keratin-19/blood , Carcinoembryonic Antigen/blood , Female , Male , Biomarkers, Tumor/blood , Prognosis , Middle Aged , Lung Neoplasms/blood , Lung Neoplasms/mortality , Lung Neoplasms/diagnosis , Lung Neoplasms/pathology , Antigens, Neoplasm/blood , Aged , Pyruvate Kinase/blood , Adult , Neoplasm Staging , Kaplan-Meier Estimate , Case-Control Studies
15.
BMJ Open ; 14(7): e083691, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39079928

ABSTRACT

INTRODUCTION: Membranopathies encompass haemolytic disorders arising from genetic variants in erythrocyte membrane proteins, including hereditary spherocytosis and stomatocytosis. Congenital dyserythropoietic anaemia type II (CDA II) is associated with the SEC23B gene and can exhibit phenotypic similarities to membranopathies. Current treatment options for these conditions, apart from splenectomy, are primarily supportive. Mitapivat, a novel pyruvate kinase (PK) activator, has demonstrated efficacy in increasing haemoglobin levels and reducing haemolysis in patients with PK deficiency, thalassemia, sickle cell disease and a mouse model of hereditary spherocytosis. METHODS AND ANALYSES: Safety and efficacy of mitapivat sulfate in adult patients with erythrocyte membranopathies (SATISFY) is a prospective, multicentre, single-arm phase two trial involving approximately 25 adult patients (≥18 years) diagnosed with a membranopathy or CDA II. During the 8-week dose escalation period, subjects will receive an initial dose of 50 mg mitapivat two times per day and may increase to 100 mg two times per day at week 4 based on the safety and changes in haemoglobin levels. Patients tolerating mitapivat well may be eligible to continue in two consecutive 24-week fixed dose periods.The primary objective of this study is to evaluate the safety of mitapivat, assessed through the occurrence of treatment-emergent adverse events. Secondary objectives include assessing the effects of mitapivat on haemoglobin levels, haemolysis, erythropoiesis, patient-reported outcome measures and spleen size.SATISFY aims to assess the safety and efficacy of mitapivat in adult patients with red blood cell membranopathies and CDA II, with the aim of establishing proof-of-concept in patients living with these rare conditions. ETHICS AND DISSEMINATION: NCT05935202/CTIS:2023-503271-24-01. Findings will be published in peer-reviewed journals. TRIAL REGISTRATION NUMBER: Clinicaltrials.gov, NCT05935202. CTIS:2023-503271-24-01. Registered 07-July-2023. Protocol number: 2.1. https://clinicaltrials.gov/study/NCT05935202.


Subject(s)
Pyruvate Kinase , Humans , Adult , Pilot Projects , Prospective Studies , Pyruvate Kinase/deficiency , Anemia, Dyserythropoietic, Congenital/drug therapy , Clinical Trials, Phase II as Topic , Pyruvate Metabolism, Inborn Errors/drug therapy , Male , Female , Multicenter Studies as Topic , Anemia, Hemolytic, Congenital Nonspherocytic
16.
J Transl Med ; 22(1): 656, 2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39004743

ABSTRACT

NAD-dependent deacetylase Sirt2 is involved in mammalian metabolic activities, matching energy demand with energy production and expenditure, and is relevant to a variety of metabolic diseases. Here, we constructed Sirt2 knockout and adeno-associated virus overexpression mice and found that deletion of hepatic Sirt2 accelerated primary obesity and insulin resistance in mice with concomitant hepatic metabolic dysfunction. However, the key targets of Sirt2 are unknown. We identified the M2 isoform of pyruvate kinase (PKM2) as a key Sirt2 target involved in glycolysis in metabolic stress. Through yeast two-hybrid and mass spectrometry combined with multi-omics analysis, we identified candidate acetylation modification targets of Sirt2 on PKM2 lysine 135 (K135). The Sirt2-mediated deacetylation-ubiquitination switch of PKM2 regulated the development of glycolysis. Here, we found that Sirt2 deficiency led to impaired glucose tolerance and insulin resistance and induced primary obesity. Sirt2 severely disrupted liver function in mice under metabolic stress, exacerbated the metabolic burden on the liver, and affected glucose metabolism. Sirt2 underwent acetylation modification of lysine 135 of PKM2 through a histidine 187 enzyme active site-dependent effect and reduced ubiquitination of the K48 ubiquitin chain of PKM2. Our findings reveal that the hepatic glucose metabolism links nutrient state to whole-body energetics through the rhythmic regulation of Sirt2.


Subject(s)
Liver , Pyruvate Kinase , Sirtuin 2 , Stress, Physiological , Ubiquitination , Animals , Humans , Male , Mice , Acetylation , Glucose/metabolism , Glycolysis , Insulin Resistance , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Pyruvate Kinase/metabolism , Sirtuin 2/metabolism
17.
Bioorg Med Chem Lett ; 110: 129865, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38950758

ABSTRACT

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.


Subject(s)
High-Throughput Screening Assays , Pyruvate Kinase , Pyruvate Kinase/metabolism , Pyruvate Kinase/antagonists & inhibitors , Humans , Drug Discovery , Structure-Activity Relationship , Urea/chemistry , Urea/pharmacology , Enzyme Activators/pharmacology , Enzyme Activators/chemistry , Enzyme Activators/chemical synthesis , Molecular Structure , Animals
18.
Biochim Biophys Acta Mol Cell Res ; 1871(7): 119796, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39038610

ABSTRACT

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.


Subject(s)
Inositol 1,4,5-Trisphosphate Receptors , Membrane Proteins , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , HeLa Cells , Membrane Proteins/metabolism , Membrane Proteins/genetics , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics , Calcium Signaling , HSP70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , Calcium/metabolism , Protein Binding , Protein Multimerization
19.
Chem Res Toxicol ; 37(8): 1394-1403, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39066737

ABSTRACT

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.


Subject(s)
Apoptosis , Pyruvate Kinase , Saponins , Cell Line, Tumor , Pyruvate Kinase/antagonists & inhibitors , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Saponins/pharmacology , Saponins/toxicity , Enzyme Activation/drug effects , Homeostasis/drug effects , Mitochondria/drug effects , Apoptosis/drug effects , Fibrosarcoma , Humans
20.
J Mol Biol ; 436(18): 168708, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39009072

ABSTRACT

Nucleoside triphosphates are indispensable in numerous biological processes, with enzymes involved in their biogenesis playing pivotal roles in cell proliferation. Pyruvate kinase (PYK), commonly regarded as the terminal glycolytic enzyme that generates ATP in tandem with pyruvate, is also capable of synthesizing a wide range of nucleoside triphosphates from their diphosphate precursors. Despite their substrate promiscuity, some PYKs show preference towards specific nucleotides, suggesting an underlying mechanism for differentiating nucleotide bases. However, the thorough characterization of this mechanism has been hindered by the paucity of nucleotide-bound PYK structures. Here, we present crystal structures of Streptococcus pneumoniae PYK in complex with four different nucleotides. These structures facilitate direct comparison of the protein-nucleotide interactions and offer structural insights into its pronounced selectivity for GTP synthesis. Notably, this selectivity is dependent on a sequence motif in the nucleotide recognition site that is widely present among prokaryotic PYKs, particularly in Firmicutes species. We show that pneumococcal cell growth is significantly impaired when expressing a PYK variant with compromised GTP and UTP synthesis activity, underscoring the importance of PYK in maintaining nucleotide homeostasis. Our findings collectively advance our understanding of PYK biochemistry and prokaryotic metabolism.


Subject(s)
Guanosine Triphosphate , Models, Molecular , Nucleotides , Pyruvate Kinase , Streptococcus pneumoniae , Pyruvate Kinase/metabolism , Pyruvate Kinase/chemistry , Pyruvate Kinase/genetics , Streptococcus pneumoniae/enzymology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Substrate Specificity , Crystallography, X-Ray , Nucleotides/metabolism , Guanosine Triphosphate/metabolism , Protein Conformation , Binding Sites , Protein Binding , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL