Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 521
Filter
1.
Life Sci ; 351: 122837, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38879156

ABSTRACT

AIM: Chronic sympathetic stimulation has been identified as a primary factor in the pathogenesis of cardiac hypertrophy (CH). However, there is no appropriate treatment available for the management of CH. Recently, it has been revealed that pyruvate kinase M2 (PKM2) plays a significant role in cardiac remodeling, fibrosis, and hypertrophy. However, the therapeutic potential of selective PKM2 inhibitor has not yet been explored in cardiac hypertrophy. Thus, in the current study, we have studied the cardioprotective potential of Compound 3K, a selective PKM2 inhibitor in isoproterenol-induced CH model. METHODS: To induce cardiac hypertrophy, male Wistar rats were subcutaneously administered isoproterenol (ISO, 5 mg/kg/day) for 14 days. Compound 3K at dosages of 2 and 4 mg/kg orally was administered to ISO-treated rats for 14 days to explore its effects on various parameters like ECG, ventricular functions, hypertrophic markers, histology, inflammation, and protein expression were performed. RESULTS: Fourteen days administration of ISO resulted in the induction of CH, which was evidenced by alterations in ECG, ventricular dysfunctions, increase in hypertrophy markers, and fibrosis. The immunoblotting of hypertrophy heart revealed the significant rise in PKM2 and reduction in PKM1 protein expression. Treatment with Compound 3K led to downregulation of PKM2 and upregulation of PKM1 protein expression. Compound 3K showed cardioprotective effects by improving ECG, cardiac functions, hypertrophy markers, inflammation, and fibrosis. Further, it also reduced cardiac expression of PKM2-associated splicing protein, HIF-1α, and caspase-3. CONCLUSION: Our findings suggest that Compound 3K has a potential cardioprotective effect via PKM2 inhibition in isoproterenol-induced CH.


Subject(s)
Cardiomegaly , Isoproterenol , Pyruvate Kinase , Animals , Male , Rats , Cardiomegaly/chemically induced , Cardiomegaly/drug therapy , Cardiomegaly/prevention & control , Cardiomegaly/metabolism , Cardiotonic Agents/pharmacology , Fibrosis , Isoproterenol/toxicity , Pyruvate Kinase/metabolism , Pyruvate Kinase/antagonists & inhibitors , Rats, Wistar , Signal Transduction/drug effects
2.
Eur J Med Chem ; 272: 116426, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38718622

ABSTRACT

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.


Subject(s)
Colitis, Ulcerative , Drug Design , Lactones , Pyruvate Kinase , Sesquiterpenes , Sesquiterpenes/pharmacology , Sesquiterpenes/chemistry , Sesquiterpenes/chemical synthesis , Animals , Mice , Pyruvate Kinase/metabolism , Pyruvate Kinase/antagonists & inhibitors , Lactones/pharmacology , Lactones/chemistry , Lactones/chemical synthesis , Structure-Activity Relationship , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/chemically induced , Humans , Molecular Structure , Cell Proliferation/drug effects , Mice, Inbred C57BL , Dose-Response Relationship, Drug , Male , Dextran Sulfate , Molecular Docking Simulation , Thyroid Hormones/metabolism , Th17 Cells/drug effects , Thyroid Hormone-Binding Proteins
3.
Bioorg Chem ; 147: 107425, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38714117

ABSTRACT

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.


Subject(s)
Carcinoma, Hepatocellular , Drug Design , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Protein Kinase Inhibitors , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Non-alcoholic Fatty Liver Disease/drug therapy , Structure-Activity Relationship , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Hep G2 Cells , Molecular Structure , Pyruvate Kinase/antagonists & inhibitors , Pyruvate Kinase/metabolism , Molecular Docking Simulation , Dose-Response Relationship, Drug , Cell Survival/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry
4.
Toxicol Appl Pharmacol ; 485: 116905, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38521371

ABSTRACT

Myocardial infarction (MI) or heart attack arises from acute or chronic prolonged ischemic conditions in the myocardium. Although several risk factors are associated with MI pathophysiology, one of the risk factors is an imbalance in the oxygen supply. The current available MI therapies are still inadequate due to the complexity of MI pathophysiology. Pyruvate kinase M2 (PKM2) has been implicated in numerous CVDs pathologies. However, the effect of specific pharmacological intervention targeting PKM2 has not been studied in MI. Therefore, in this study, we explored the effect of compound 3K, a PKM2-specific inhibitor, in isoproterenol-induced acute MI model. In this study, in order to induce MI in rats, isoproterenol (ISO) was administered at a dose of 100 mg/kg over two days at an interval of 24 h. Specific PKM2 inhibitor, compound 3K (2 and 4 mg/kg), was administered in MI rats to investigate its cardioprotective potential. After the last administration of compound 3K, ECG and hemodynamic parameters were recorded using a PV-loop system. Cardiac histology, western blotting, and plasmatic cardiac damage markers were evaluated to elucidate the underlying mechanisms. Treatment of compound 3K significantly reduced ISO-induced alterations in ECG, ventricular functions, cardiac damage, infarct size, and cardiac fibrosis. Compound 3K treatment produced significant increase in PKM1 expression and decrease in PKM2 expression. In addition, HIF-1α, caspase-3, c-Myc, and PTBP1 expression were also reduced after compound 3K treatment. This study demonstrates the cardioprotective potential of compound 3K in MI, and its mechanisms of cardioprotective action.


Subject(s)
Cardiotonic Agents , Isoproterenol , Myocardial Infarction , Pyruvate Kinase , Animals , Isoproterenol/toxicity , Myocardial Infarction/chemically induced , Myocardial Infarction/prevention & control , Myocardial Infarction/pathology , Male , Rats , Pyruvate Kinase/metabolism , Pyruvate Kinase/antagonists & inhibitors , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Rats, Wistar , Myocardium/pathology , Myocardium/metabolism , Myocardium/enzymology , Disease Models, Animal , Rats, Sprague-Dawley , Protein Kinase Inhibitors/pharmacology , Thyroid Hormones
5.
Mol Metab ; 66: 101625, 2022 12.
Article in English | MEDLINE | ID: mdl-36374631

ABSTRACT

OBJECTIVE: The endocrine pancreatic ß-cells play a pivotal role in maintaining whole-body glucose homeostasis and its dysregulation is a consistent feature in all forms of diabetes. However, knowledge of intracellular regulators that modulate ß-cell function remains incomplete. We investigated the physiological role of ROCK1 in the regulation of insulin secretion and glucose homeostasis. METHODS: Mice lacking ROCK1 in pancreatic ß-cells (RIP-Cre; ROCK1loxP/loxP, ß-ROCK1-/-) were studied. Glucose and insulin tolerance tests as well as glucose-stimulated insulin secretion (GSIS) were measured. An insulin secretion response to a direct glucose or pyruvate or pyruvate kinase (PK) activator stimulation in isolated islets from ß-ROCK1-/- mice or ß-cell lines with knockdown of ROCK1 was also evaluated. A proximity ligation assay was performed to determine the physical interactions between PK and ROCK1. RESULTS: Mice with a deficiency of ROCK1 in pancreatic ß-cells exhibited significantly increased blood glucose levels and reduced serum insulin without changes in body weight. Interestingly, ß-ROCK1-/- mice displayed a progressive impairment of glucose tolerance while maintaining insulin sensitivity mostly due to impaired GSIS. Consistently, GSIS markedly decreased in ROCK1-deficient islets and ROCK1 knockdown INS-1 cells. Concurrently, ROCK1 blockade led to a significant decrease in intracellular calcium and ATP levels and oxygen consumption rates in isolated islets and INS-1 cells. Treatment of ROCK1-deficient islets or ROCK1 knockdown ß-cells either with pyruvate or a PK activator rescued the impaired GSIS. Mechanistically, we observed that glucose stimulation in ß-cells greatly enhanced ROCK1 binding to PK. CONCLUSIONS: Our findings demonstrate that ß-cell ROCK1 is essential for glucose-stimulated insulin secretion and for glucose homeostasis and that ROCK1 acts as an upstream regulator of glycolytic pyruvate kinase signaling.


Subject(s)
Insulin Secretion , Insulin , Pyruvate Kinase , rho-Associated Kinases , Animals , Mice , Glucose/metabolism , Insulin/metabolism , Insulin Secretion/physiology , Pyruvate Kinase/antagonists & inhibitors , Pyruvate Kinase/metabolism , Pyruvates
6.
Biochim Biophys Acta Mol Cell Res ; 1869(4): 119206, 2022 04.
Article in English | MEDLINE | ID: mdl-35026348

ABSTRACT

Pyruvate kinase isoform M2 (PKM2) is a rate-limiting glycolytic enzyme that is widely expressed in embryonic tissues. The expression of PKM2 declines in some tissues following embryogenesis, while other pyruvate kinase isozymes are upregulated. However, PKM2 is highly expressed in cancer cells and is believed to play a role in supporting anabolic processes during tumour formation. In this study, PKM2 was identified as an inositol 1,4,5-trisphosphate receptor (IP3R)-interacting protein by mass spectrometry. The PKM2:IP3R interaction was further characterized by pull-down and co-immunoprecipitation assays, which showed that PKM2 interacted with all three IP3R isoforms. Moreover, fluorescence microscopy indicated that both IP3R and PKM2 localized at the endoplasmic reticulum. PKM2 binds to IP3R at a highly conserved 21-amino acid site (corresponding to amino acids 2078-2098 in mouse type 1 IP3R isoform). Synthetic peptides (denoted 'TAT-D5SD' and 'D5SD'), based on the amino acid sequence at this site, disrupted the PKM2:IP3R interaction and potentiated IP3R-mediated Ca2+ release both in intact cells (TAT-D5SD peptide) and in a unidirectional 45Ca2+ flux assay on permeabilized cells (D5SD peptide). The TAT-D5SD peptide did not affect the enzymatic activity of PKM2. Reducing PKM2 protein expression using siRNA increased IP3R-mediated Ca2+ signalling in intact cells without altering the ER Ca2+ content. These data identify PKM2 as an IP3R-interacting protein that inhibits intracellular Ca2+ signalling. The elevated expression of PKM2 in cancer cells is therefore not solely connected to its canonical role in glycolytic metabolism, rather PKM2 also has a novel non-canonical role in regulating intracellular signalling.


Subject(s)
Calcium Signaling , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Pyruvate Kinase/metabolism , Amino Acid Sequence , Animals , Calcium/metabolism , Cell Line , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Lymphocytes/cytology , Lymphocytes/metabolism , Mice , Protein Binding , Protein Domains , Protein Isoforms/metabolism , Pyruvate Kinase/antagonists & inhibitors , Pyruvate Kinase/genetics , RNA Interference , RNA, Small Interfering/metabolism
7.
Sci Rep ; 11(1): 21726, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34741111

ABSTRACT

Cryptotanshinone (CT) is an extract from the traditional Chinese medicine Salvia miltiorrhiza, which inhibits the growth of methicillin-resistant Staphylococcus aureus (MRSA) in vitro. This study aims to determine the antibacterial mechanisms of CT by integrating bioinformatics analysis and microbiology assay. The microarray data of GSE13203 was retrieved from the Gene Expression Omnibus (GEO) database to screen the differentially expressed genes (DEGs) of S. aureus strains that were treated with CT treatment. Gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were used to identify the potential target of CT. Data mining on the microarray dataset indicated that pyruvate kinase (PK) might be involved in the antimicrobial activities of CT. The minimum inhibition concentrations (MICs) of CT or vancomycin against the MRSA strain ATCC43300 and seven other clinical strains were determined using the broth dilution method. The effects of CT on the activity of PK were further measured. In vitro tests verified that CT inhibited the growth of an MRSA reference strain and seven other clinical strains. CT hampered the activity of the PK of ATCC43300 and five clinical MRSA strains. CT might hinder bacterial energy metabolism by inhibiting the activity of PK.


Subject(s)
Methicillin-Resistant Staphylococcus aureus/drug effects , Phenanthrenes/pharmacology , Pyruvate Kinase/antagonists & inhibitors , Computational Biology , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Gene Expression Profiling , Humans , Methicillin-Resistant Staphylococcus aureus/enzymology , Phenanthrenes/therapeutic use , Phytotherapy , Staphylococcal Infections/drug therapy
8.
Exp Eye Res ; 213: 108823, 2021 12.
Article in English | MEDLINE | ID: mdl-34752817

ABSTRACT

Choroidal neovascularization (CNV), a feature of neovasular age-related macular degeneration (AMD), acts as a leading cause of vision loss in the elderly. Shikonin (SHI), a natural bioactive compound extracted from Chinese herb radix arnebiae, exerts anti-inflammatory and anti-angiogenic roles and also acts as a potential pyruvate kinase M2 (PKM2) inhibitor in macrophages. The major immune cells macrophages infiltrate the CNV lesions, where the production of pro-angiognic cytokines from macrophage facilitates the development of CNV. PKM2 contributes to the neovascular diseases. In this study, we found that SHI oral gavage alleviated the leakage, area and volume of mouse laser-induced CNV lesion and inhibited macrophage infiltration without ocular cytotoxicity. Moreover, SHI inhibited the secretion of pro-angiogenic cytokine, including basic fibroblast growth factor (FGF2), insulin-like growth factor-1 (IGF1), chemokine (C-C motif) ligand 2 (CCL2), placental growth factor and vascular endothelial growth factor (VEGF), from primary human macrophages by down-regulating PKM2/STAT3/CD163 pathway, indicating a novel potential therapy strategy for CNV.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Choroidal Neovascularization/drug therapy , Macrophages/drug effects , Naphthoquinones/therapeutic use , Pyruvate Kinase/antagonists & inhibitors , Angiogenesis Inducing Agents/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Blotting, Western , Cells, Cultured , Choroidal Neovascularization/enzymology , Chromatography, High Pressure Liquid , Coloring Agents/administration & dosage , Cytokines/metabolism , Disease Models, Animal , Drugs, Chinese Herbal/therapeutic use , Enzyme-Linked Immunosorbent Assay , Fluorescein Angiography , Humans , In Situ Nick-End Labeling , Indocyanine Green/administration & dosage , Macrophages/enzymology , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Pyruvate Kinase/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism
9.
ACS Appl Mater Interfaces ; 13(48): 56972-56987, 2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34797638

ABSTRACT

Triple-negative breast cancer (TNBC) is typically associated with poor prognosis due to its only partial response to chemotherapy and lack of clinically established targeted therapies coupled with an aggressive disease course. Aerobic glycolysis is a hallmark of reprogrammed metabolic activity in cancer cells, which can be repressed by small-interfering RNA (siRNA). However, the lack of effective carriers to deliver vulnerable siRNA restricts the clinical potentials of glycolysis-based gene therapy for TNBC. Herein, we develop a tumor-targeted, biomimetic manganese dioxide (MnO2)-shrouded metal-organic framework (MOF) based nanomedicine to deliver siRNA against pyruvate kinase muscle isozyme M2 (siPKM2), wherein PKM2 is a rate-limiting enzyme in glycolysis, to inhibit the reprogrammed glycolysis of TNBC. This MOF-based genetic nanomedicine shows excellent monodispersity and stability and protects siPKM2 against degradation by nucleases. The nanomedicine not only substantially blocks the glycolytic pathway but also improves intracellular hypoxia in TNBC cells, with a resultant O2-enhanced anticancer effect. In the mice orthotopic TNBC model, the nanomedicine shows a remarkable therapeutic effect. Meanwhile, the Mn2+ ions released from acid microenvironment-responsive MnO2 enable in vivo monitoring of the therapeutic process with magnetic resonance imaging (MRI). Our study shows great promise with this MRI-visible MOF-based nanomedicine for treating TNBC by inhibition of glycolysis via the RNA interference.


Subject(s)
Antineoplastic Agents/pharmacology , Biomimetic Materials/pharmacology , Enzyme Inhibitors/pharmacology , Metal-Organic Frameworks/pharmacology , Pyruvate Kinase/antagonists & inhibitors , Theranostic Nanomedicine , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Biomimetic Materials/chemical synthesis , Biomimetic Materials/chemistry , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Female , Gene Silencing/drug effects , Humans , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Materials Testing , Metal-Organic Frameworks/chemical synthesis , Metal-Organic Frameworks/chemistry , Mice , Mice, Nude , Optical Imaging , Oxides/chemistry , Oxides/pharmacology , Particle Size , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Triple Negative Breast Neoplasms/diagnostic imaging , Triple Negative Breast Neoplasms/metabolism
10.
Dis Markers ; 2021: 1602797, 2021.
Article in English | MEDLINE | ID: mdl-34804260

ABSTRACT

Retinitis pigmentosa (RP) is a major cause of blindness that is difficult to diagnose and treat. PKM2, a subtype of pyruvate kinase, is strongly associated with oxidative stress and is expressed in photoreceptors. We investigated whether PKM2 reduces photoreceptor cell apoptosis and evaluated possible antiapoptotic mechanisms in RP. We established RP models by exposing 661W cells to blue light and modulated PKM2 activity using a PKM2 inhibitor. We measured the apoptosis rates using calcein-acetoxymethyl ester/propidium iodide double staining and Cell Counting Kit-8, the oxidative stress levels using a reactive oxygen species assay, and the changes in protein expression by western blotting. Photodamage increased PKM2 expression, cellular oxidative stress, and apoptosis of 661W cells. PKM2 inhibition significantly reduced the levels of apoptosis and oxidative stress induced by photodamage. Our data suggest that PKM2 is a potential disease marker and therapeutic target for RP.


Subject(s)
Light/adverse effects , Neuroprotection , Oxidative Stress , Photoreceptor Cells/metabolism , Pyruvate Kinase/antagonists & inhibitors , Retinal Cone Photoreceptor Cells/metabolism , Retinitis Pigmentosa/prevention & control , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Mice , Photoreceptor Cells/pathology , Photoreceptor Cells/radiation effects , Reactive Oxygen Species/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Cone Photoreceptor Cells/radiation effects , Retinitis Pigmentosa/etiology , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology
11.
J Cell Mol Med ; 25(22): 10711-10723, 2021 11.
Article in English | MEDLINE | ID: mdl-34687136

ABSTRACT

Hypertensive cardiac remodelling is a common cause of heart failure. However, the molecular mechanisms regulating cardiac remodelling remain unclear. Pyruvate kinase isozyme type M2 (PKM2) is a key regulator of the processes of glycolysis and oxidative phosphorylation, but the roles in cardiac remodelling remain unknown. In the present study, we found that PKM2 was enhanced in angiotensin II (Ang II)-treated cardiac fibroblasts and hypertensive mouse hearts. Suppression of PKM2 by shikonin alleviated cardiomyocyte hypertrophy and fibrosis in Ang-II-induced cardiac remodelling in vivo. Furthermore, inhibition of PKM2 markedly attenuated the function of cardiac fibroblasts including proliferation, migration and collagen synthesis in vitro. Mechanistically, suppression of PKM2 inhibited cardiac remodelling by suppressing TGF-ß/Smad2/3, Jak2/Stat3 signalling pathways and oxidative stress. Together, this study suggests that PKM2 is an aggravator in Ang-II-mediated cardiac remodelling. The negative modulation of PKM2 may provide a promising therapeutic approach for hypertensive cardiac remodelling.


Subject(s)
Angiotensin II/metabolism , Janus Kinase 2/metabolism , Oxidative Stress/drug effects , Pyruvate Kinase/genetics , STAT3 Transcription Factor/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Ventricular Remodeling/genetics , Animals , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression , Hypertension/complications , Hypertension/etiology , Hypertension/metabolism , Male , Mice , Models, Biological , Pyruvate Kinase/antagonists & inhibitors , Reactive Oxygen Species/metabolism
12.
Curr Top Med Chem ; 21(25): 2258-2271, 2021.
Article in English | MEDLINE | ID: mdl-34348626

ABSTRACT

INTRODUCTION: Pyruvate kinase isozyme M2 (PKM2) was observed to be overexpressed and play a key role in cell growth and cancer cells' metabolism. During the past years, phytochemicals have been developed as new treatment options for chemoprevention and cancer therapy. Natural resources, like shikonin (naphthoquinone) and its derivatives, have emerged to be high potential therapeutics in cancer treatment. METHODS: Our study aimed to design novel anti-tumour agents (PKM2 inhibitors) focusing on the shikonin scaffold with a better activity using computational methods. We applied a three-dimensional quantitative structure-activity relationship (3D-QSAR) approach using Field-based QSAR. RESULTS: The Comparative Molecular Field Analysis (CoMFA) and Comparative Molecular Similarity Indices Analysis (CoMSIA) were performed on a series of forty shikonin derivatives, including shikonin, to develop robust models and rationalize the PKM2 inhibitory activity profile by building a correlation between structural features and activity. CONCLUSION: These predictive computational models will further help the design and synthesis of potent PKM2 inhibitors and their fast biological assessment at a low cost.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Naphthoquinones/chemistry , Pyruvate Kinase/antagonists & inhibitors , Quantitative Structure-Activity Relationship , Isoenzymes/antagonists & inhibitors
13.
Int J Biol Sci ; 17(8): 1895-1908, 2021.
Article in English | MEDLINE | ID: mdl-34131394

ABSTRACT

Ovarian cancer is a common cause of death among gynecological cancers. Although ovarian cancer initially responds to chemotherapy, frequent recurrence in patients remains a therapeutic challenge. Pyruvate kinase M2 (PKM2) plays a pivotal role in regulating cancer cell survival. However, its therapeutic role remains unclear. Here, we investigated the anticancer effects of compound 3K, a specific PKM2 inhibitor, on the regulation of autophagic and apoptotic pathways in SK-OV-3 (PKM2-overexpressing human ovarian adenocarcinoma cell line). The anticancer effect of compound 3K was examined using MTT and colony formation assays in SK-OV-3 cells. PKM2 expression was positively correlated with the severity of the tumor, and expression of pro-apoptotic proteins increased in SK-OV-3 cells following compound 3K treatment. Compound 3K induced AMPK activation, which was accompanied by mTOR inhibition. Additionally, this compound inhibited glycolysis, resulting in reduced proliferation of SK-OV-3 cells. Compound 3K treatment suppressed tumor progression in an in vivo xenograft model. Our findings suggest that the inhibition of PKM2 by compound 3K affected the Warburg effect and induced autophagic cell death. Therefore, use of specific PKM2 inhibitors to block the glycolytic pathway and target cancer cell metabolism represents a promising therapeutic approach for treating PKM2-overexpressing ovarian cancer.


Subject(s)
Adenocarcinoma , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Ovarian Neoplasms , Pyruvate Kinase/antagonists & inhibitors , Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , Autophagic Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Signal Transduction/drug effects , Tumor Stem Cell Assay
14.
J Med Chem ; 63(21): 12623-12641, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33103423

ABSTRACT

Natural bisindole alkaloids such as Hyrtinadine A and Alocasin A, which are known to exhibit diverse bioactivities, provide promising chemical scaffolds for drug development. By optimizing the Masuda borylation-Suzuki coupling sequence, a library of various natural product-derived and non-natural (di)azine-bridged bisindoles was created. While unsubstituted bisindoles were devoid of antibacterial activity, 5,5'-chloro derivatives were highly active against methicillin-resistant Staphylococcus aureus (MRSA) and further Gram-positive pathogens at minimal inhibitory concentrations ranging from 0.20 to 0.78 µM. These compounds showed strong bactericidal killing effects but only moderate cytotoxicity against human cell lines. Furthermore, the two front-runner compounds 4j and 4n exhibited potent in vivo efficacy against MRSA in a mouse wound infection model. Although structurally related bisindoles were reported to specifically target pyruvate kinase in MRSA, antibacterial activity of 4j and 4n is independent of pyruvate kinase. Rather, these compounds lead to bacterial membrane permeabilization and cellular efflux of low-molecular-weight molecules.


Subject(s)
Alkaloids/chemistry , Anti-Bacterial Agents/pharmacology , Indoles/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Alkaloids/metabolism , Alkaloids/pharmacology , Alkaloids/therapeutic use , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/therapeutic use , Cell Line , Cell Survival/drug effects , Female , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Humans , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Peritonitis/drug therapy , Peritonitis/pathology , Pyruvate Kinase/antagonists & inhibitors , Pyruvate Kinase/metabolism , Rabbits
15.
Elife ; 92020 08 19.
Article in English | MEDLINE | ID: mdl-32812866

ABSTRACT

Natural Killer (NK) cells have an important role in immune responses to viruses and tumours. Integrating changes in signal transduction pathways and cellular metabolism is essential for effective NK cells responses. The glycolytic enzyme Pyruvate Kinase Muscle 2 (PKM2) has described roles in regulating glycolytic flux and signal transduction, particularly gene transcription. While PKM2 expression is robustly induced in activated NK cells, mice lacking PKM2 in NK cells showed no defect in NK cell metabolism, transcription or antiviral responses to MCMV infection. NK cell metabolism was maintained due to compensatory PKM1 expression in PKM2-null NK cells. To further investigate the role of PKM2, we used TEPP-46, which increases PKM2 catalytic activity while inhibiting any PKM2 signalling functions. NK cells activated with TEPP-46 had reduced effector function due to TEPP-46-induced increases in oxidative stress. Overall, PKM2-regulated glycolytic metabolism and redox status, not transcriptional control, facilitate optimal NK cells responses.


Subject(s)
Gene Expression Regulation , Glycolysis , Killer Cells, Natural/metabolism , Pyruvate Kinase , Animals , Cells, Cultured , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glycolysis/drug effects , Glycolysis/genetics , Mice , Oxidative Stress , Pyridazines/pharmacology , Pyrroles/pharmacology , Pyruvate Kinase/antagonists & inhibitors , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Signal Transduction
16.
JCI Insight ; 5(17)2020 09 03.
Article in English | MEDLINE | ID: mdl-32750036

ABSTRACT

One of the most significant adverse postburn responses is abnormal scar formation, such as keloids. Despite its prolificacy, the underlying pathophysiology of keloid development is unknown. We recently demonstrated that NLRP3 inflammasome, the master regulator of inflammatory and metabolic responses (e.g., aerobic glycolysis), is essential for physiological wound healing. Therefore, burn patients who develop keloids may exhibit altered immunometabolic responses at the site of injury, which interferes with normal healing and portends keloid development. Here, we confirmed keloid NLRP3 activation (cleaved caspase-1 [P < 0.05], IL-1ß [P < 0.05], IL-18 [P < 0.01]) and upregulation in Glut1 (P < 0.001) and glycolytic enzymes. Burn skin similarly displayed enhanced glycolysis and Glut1 expression (P < 0.01). However, Glut1 was significantly higher in keloid compared with nonkeloid burn patients (>2 SD above mean). Targeting aberrant glucose metabolism with shikonin, a pyruvate kinase M2 inhibitor, dampened NLRP3-mediated inflammation (cleaved caspase-1 [P < 0.05], IL-1ß [P < 0.01]) and improved healing in vivo. In summary, burn skin exhibited evidence of Warburg-like metabolism, similar to keloids. Targeting this altered metabolism could change the trajectory toward normal scarring, indicating the clinical possibility of shikonin for abnormal scar prevention.


Subject(s)
Burns/complications , Glycolysis , Inflammation/prevention & control , Keloid/prevention & control , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Skin/drug effects , Wound Healing , Adult , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Case-Control Studies , Female , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Humans , Inflammasomes , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators , Keloid/etiology , Keloid/metabolism , Keloid/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Naphthoquinones/pharmacology , Pyruvate Kinase/antagonists & inhibitors
17.
Biochem Biophys Res Commun ; 528(3): 594-600, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32507600

ABSTRACT

Pyruvate kinase M2 (PKM2) functions as an important rate-limiting enzyme of aerobic glycolysis that is involved in tumor initiation and progression. However, there are few studies on effective PKM2 inhibitors. Gliotoxin is a marine-derived fungal secondary metabolite with multiple biological activities, including immunosuppression, cytotoxicity, and et al. In this study, we found that Gliotoxin directly bound to PKM2 and inhibited its glycolytic activity in a dose-dependent manner accompanied by the decreases in glucose consumption and lactate production in the human glioma cell line U87. Moreover, Gliotoxin suppressed tyrosine kinase activity of PKM2, leading to a dramatic reduction in Stat3 phosphorylation in U87 cells. Furthermore, Gliotoxin suppressed cell viability in U87 cells, and cytotoxicity of Gliotoxin on U87 cells was obviously augmented under hypoxia condition compared to normal condition. Finally, Gliotoxin was demonstrated to induce cell apoptosis of U87 cells and synergize with temozolomide. Our findings identify Gliotoxin as a new PKM2 inhibitor with anti-tumor activity, which lays the foundation for the development of Gliotoxin as a promising anti-tumor drug in the future.


Subject(s)
Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Gliotoxin/isolation & purification , Gliotoxin/pharmacology , Pyruvate Kinase/antagonists & inhibitors , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Aquatic Organisms/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cell-Free System , Drug Synergism , Enzyme Inhibitors/administration & dosage , Fungi/chemistry , Gliotoxin/administration & dosage , Glycolysis/drug effects , Humans , Phosphorylation , Temozolomide/administration & dosage
18.
J Immunol ; 205(1): 181-192, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32503893

ABSTRACT

Recent studies indicate that glucose metabolism is altered in rheumatoid arthritis. We hypothesize that Pkm2, as a key regulatory enzyme of glycolysis pathway, triggers the activation of macrophages (Mφ), which results in proinflammatory cytokine production during the arthritis progress. In this study, Pkm2 was found to be overexpressed in ED1-positive Mφ in spleens and synovial tissues from arthritic rats via immunofluorescence, Western blotting, and quantitative RT-PCR. To reveal the role of Pkm2, Dark Agouti rats were treated with either Pkm2 enzyme inhibitor shikonin or the RNA interference plasmids of Pkm2 and negative control plasmids, respectively, via i.p. injection. Pkm2 intervention could alleviate the severity of pristane-induced arthritis in aspects of the macroscopic arthritis score, perimeter changes of midpaw, and the synovitis and destruction of the bone and cartilage as well as reduce the ED1 and p-Stat1-positive cell population in rat synovial tissues. Silencing Pkm2 by RNA interference in classical activated rat and mouse Mφ resulted in less Tnf-α, Il-1ß production via Stat1 signaling. Collectively, Pkm2 is highly expressed in ED1-positive Mφ of spleens and synovial tissues from arthritic rats and promotes Mφ activation via Stat1 signaling. Pkm2 might be a promising selective metabolic target molecule for rheumatoid arthritis treatment.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Macrophages/immunology , Pyruvate Kinase/metabolism , STAT1 Transcription Factor/metabolism , Animals , Arthritis, Experimental/diagnosis , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/pathology , Gene Knockdown Techniques , Humans , Macrophages/metabolism , Mice , Naphthoquinones/administration & dosage , Pyruvate Kinase/antagonists & inhibitors , Pyruvate Kinase/genetics , RAW 264.7 Cells , RNA, Small Interfering/metabolism , Rats , Severity of Illness Index , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Synovial Membrane/immunology , Synovial Membrane/pathology
19.
Mar Drugs ; 18(5)2020 May 05.
Article in English | MEDLINE | ID: mdl-32380771

ABSTRACT

Liquid chromatography coupled with high resolution mass spectrometry (LC-HRESMS)-assisted metabolomic profiling of two sponge-associated actinomycetes, Micromonospora sp. UR56 and Actinokineospora sp. EG49, revealed that the co-culture of these two actinomycetes induced the accumulation of metabolites that were not traced in their axenic cultures. Dereplication suggested that phenazine-derived compounds were the main induced metabolites. Hence, following large-scale co-fermentation, the major induced metabolites were isolated and structurally characterized as the already known dimethyl phenazine-1,6-dicarboxylate (1), phenazine-1,6-dicarboxylic acid mono methyl ester (phencomycin; 2), phenazine-1-carboxylic acid (tubermycin; 3), N-(2-hydroxyphenyl)-acetamide (9), and p-anisamide (10). Subsequently, the antibacterial, antibiofilm, and cytotoxic properties of these metabolites (1-3, 9, and 10) were determined in vitro. All the tested compounds except 9 showed high to moderate antibacterial and antibiofilm activities, whereas their cytotoxic effects were modest. Testing against Staphylococcus DNA gyrase-B and pyruvate kinase as possible molecular targets together with binding mode studies showed that compounds 1-3 could exert their bacterial inhibitory activities through the inhibition of both enzymes. Moreover, their structural differences, particularly the substitution at C-1 and C-6, played a crucial role in the determination of their inhibitory spectra and potency. In conclusion, the present study highlighted that microbial co-cultivation is an efficient tool for the discovery of new antimicrobial candidates and indicated phenazines as potential lead compounds for further development as antibiotic scaffold.


Subject(s)
Actinobacteria/metabolism , Anti-Bacterial Agents/pharmacology , Micromonospora/metabolism , Porifera/microbiology , Topoisomerase II Inhibitors/pharmacology , Actinobacteria/isolation & purification , Animals , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Bacteriological Techniques/methods , Biofilms/drug effects , DNA Gyrase/metabolism , Enzyme Assays , Fermentation , Metabolomics/methods , Microbial Sensitivity Tests , Micromonospora/isolation & purification , Molecular Conformation , Molecular Docking Simulation , Pyruvate Kinase/antagonists & inhibitors , Pyruvate Kinase/metabolism , Staphylococcus/drug effects , Staphylococcus/enzymology , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/isolation & purification , Topoisomerase II Inhibitors/metabolism
20.
Pharmacol Rep ; 72(3): 631-640, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32040844

ABSTRACT

BACKGROUND: The M2 isoform of the glycolytic enzyme pyruvate kinase (PKM2) is one of the key components in the Warburg effect, and an important regulator of cancer cell metabolism. Elevated PKM2 expression is a hallmark of numerous tumor types, making it a promising target for cancer therapy. METHODS: Migration of H1299 lung tumor cells treated with synthetic oleanane triterpenoid derivatives CDDO-Me and CDDO-Im was monitored using scratch and transwell assays. Direct binding and inhibition of PKM2 activity by CDDO-Me was demonstrated by pull-down and activity assays. PKM2 localization in the absence and presence of CDDO-Me or CDDO-Im was determined by subcellular fractionation and immunofluorescence microscopy. Involvement of PKM2 in tumor cell migration was assessed using a stable PKM2 knockdown cell line. RESULTS: We demonstrate that migration of H1299 lung tumor cells is inhibited by CDDO-Me and CDDO-Im in scratch and transwell assays. CDDO-Me binds directly and specifically to recombinant PKM2, leading to a reduction of its catalytic activity. PKM2 knockdown cells exhibit significantly lower migration compared to control cells when subjected to glucose and oxygen deprivation, but not under regular conditions. CONCLUSIONS: The results suggest that PKM2 expression in a tumor-like environment contributes to cell migration, and that PKM2 activity can be down regulated by synthetic triterpenoid derivatives.


Subject(s)
Cell Movement/drug effects , Oleanolic Acid/analogs & derivatives , Pyruvate Kinase/antagonists & inhibitors , Pyruvate Kinase/metabolism , Carcinoma, Non-Small-Cell Lung , Cell Line, Tumor , Glucose , Humans , Hypoxia , Oleanolic Acid/metabolism , Oleanolic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL