Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 591
Filter
Add more filters

Publication year range
1.
Eur J Nutr ; 61(8): 4059-4075, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35804267

ABSTRACT

PURPOSE: Ferroptosis is a form of regulated cell death that has the potential to be targeted as a cancer therapeutic strategy. But cancer cells have a wide range of sensitivities to ferroptosis, which limits its therapeutic potential. Accumulation of lipid peroxides determines the occurrence of ferroptosis. However, the type of lipid involved in peroxidation and the mechanism of lipid peroxide accumulation are less studied. METHODS: The effects of fatty acids (10 µM) with different carbon chain length and unsaturation on ferroptosis were evaluated by MTT and LDH release assay in cell lines derived from prostate cancer (PC3, 22RV1, DU145 and LNCaP), colorectal cancer (HT-29), cervical cancer (HeLa) and liver cancer (HepG2). Inhibitors of apoptosis, necroptosis, autophagy and ferroptosis were used to determine the type of cell death. Then the regulation of reactive oxygen species (ROS) and lipid peroxidation by docosahexaenoic acid (DHA) was measured by HPLC-MS and flow cytometry. The avtive form of DHA was determined by siRNA mediated gene silencing. The role of lipoxygenases was checked by inhibitors and gene silencing. Finally, the effect of DHA on ferroptosis-mediated tumor killing was verified in xenografts. RESULTS: The sensitivity of ferroptosis was positively correlated with the unsaturation of exogenously added fatty acid. DHA (22:6 n-3) sensitized cancer cells to ferroptosis-inducing reagents (FINs) at the highest level in vitro and in vivo. In this process, DHA increased ROS accumulation, lipid peroxidation and protein oxidation independent of its membrane receptor, GPR120. Inhibition of long chain fatty acid-CoA ligases and lysophosphatidylcholine acyltransferases didn't affect the role of DHA. DHA-involved ferroptosis can be induced in both arachidonate lipoxygenase 5 (ALOX5) negative and positive cells. Down regulation of ALOX5 inhibited ferroptosis, while overexpression of ALOX5 promoted ferroptosis. CONCLUSION: DHA can effectively promote ferroptosis-mediated tumor killing by increasing intracellular lipid peroxidation. Both ALOX5 dependent and independent pathways are involved in DHA-FIN induced ferroptosis. And during this process, free DHA plays an important role.


Subject(s)
Docosahexaenoic Acids , Neoplasms , Male , Humans , Docosahexaenoic Acids/pharmacology , Reactive Oxygen Species/metabolism , Lipid Peroxides , Lipoxygenase/metabolism , Lipoxygenase/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Lysophosphatidylcholines/pharmacology , Cell Line, Tumor , Cell Death , Lipid Peroxidation , Lipoxygenases/metabolism , Arachidonate Lipoxygenases/metabolism , Arachidonate Lipoxygenases/pharmacology , Acyltransferases/metabolism , Acyltransferases/pharmacology , Carbon , Coenzyme A/metabolism , Coenzyme A/pharmacology
2.
Medicina (Kaunas) ; 58(9)2022 Sep 08.
Article in English | MEDLINE | ID: mdl-36143922

ABSTRACT

Background and Objectives: Methicillin-resistant Staphylococcus aureus (MRSA) isolates are associated with various diseases ranged from mild superficial impairments to invasive infections. This study aimed to evaluate the ability of polymerase chain reaction (PCR) based methods namely, restriction fragment length polymorphism (RFLP) of the coa gene and random amplified polymorphic DNA (RAPD), to determine the genetic diversity of MRSA isolates. Materials and Methods: A total of 37 MRSA isolates were conventionally identified depending on their biochemical and microbiological culture characteristics. Genotypic confirmation was based on detection of the associated mecA gene. The genetic variation amongst MRSA isolates was evaluated following the coa gene-based RFLP and RAPD fingerprints. Results: Results illustrated that, the species specific coa gene was detected in all MRSA isolates. The irregular bands intensity, number, and molecular sizes of the PCR amplicons demonstrated the coa gene polymorphism. The incompatible AluI digestion patterns of these amplicons classified the tested MRSA isolates into 20 RFLP patterns which confirm the coa gene polymorphism. Additionally, the PCR-based RAPD analysis showed variable bands number with size range of approximately 130 bp to 4 kbp, which indicated the genetic variation of the tested MRSA isolates as it created 36 variable RAPD banding profiles. Conclusions: coa gene AluI enzymatic restriction sites, amongst the tested MRSA isolates, certify their genetic variation on the basis of the accurate but complicated and relatively expensive coa gene-based RFLP. Conversely, the results verified the excellent ability of the simple and cost-effective PCR-based RAPD analysis to discriminate between MRSA isolates without any preface data about the genome.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Coagulase/genetics , Coagulase/pharmacology , Coenzyme A/genetics , Coenzyme A/pharmacology , DNA/pharmacology , Humans , Methicillin-Resistant Staphylococcus aureus/genetics , Polymorphism, Restriction Fragment Length , Random Amplified Polymorphic DNA Technique , Staphylococcal Infections/diagnosis , Staphylococcus aureus/genetics
3.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 53(5): 805-814, 2022 Sep.
Article in Zh | MEDLINE | ID: mdl-36224682

ABSTRACT

Objective: To explore the effects of hydroxyacyl-CoA dehydrogenase alpha subunit (HADHA) on the migration and invasion of HTR-8/SVneo cells, a human trophoblast cell line, and its potential mechanism of action. Methods: Immunofluorescence staining was done to evaluate the expression levels of HADHA in samples of normal villi and recurrent spontaneous abortion (RSA) villi at 6-8 weeks. Lentiviral infection system was used to construct stable HTR-8/SVneo cell lines with HADHA overexpression and knockdown. Western blot, qRT-PCR, Wound-healing assay, and Transwell assay were used to determine the effect of HADHA on the migration and invasion of HTR-8/SVneo cells and the expression of relevant genes. Transcriptome sequencing and bioinformatics analysis were done to screen for the potential target genes and signaling pathways regulated by HADHA. The specific molecular mechanism of how HADHA regulates the migration and invasion of HTR-8/SVneo cells was examined by adding the inhibitor of protein kinase B (PKB/AKT). Results: HADHA was highly expressed in extravillous trophoblasts (EVT) of RSA villus samples as compared with samples from the normal control group. In HTR-8/SVneo cells overexpressing HADHA, the expression levels of migration and invasion-related genes, including HLA-G, MMP2, MMP9, and NCAD, were decreased (P<0.01,P<0.05), and the migration and invasion abilities of HTR-8/SVneo cells were weakened (P<0.05). HADHA knockdown increased the expression levels of HLA-G, MMP2, MMP9, and NCAD (P<0.01, P<0.05), and promoted the migration and invasion of HTR-8/SVneo cells (P<0.05). In addition, HADHA overexpression decreased the phosphorylation levels of PI3K and AKT (P<0.05) and inhibited the PI3K/AKT signaling pathway. HADHA knockdown activated the PI3K/AKT signaling pathway. When MK-2206, an AKT inhibitor, was added to stable HTR-8/SVneo cell lines with HADHA knockdown, the migration and invasion of the cells were significantly reduced. Conclusion: HADHA inhibits the migration and invasion of HTR-8/SVneo cells by inhibiting the PI3K/AKT signaling pathway.


Subject(s)
Pre-Eclampsia , Proto-Oncogene Proteins c-akt , Cell Movement/physiology , Coenzyme A/metabolism , Coenzyme A/pharmacology , Female , HLA-G Antigens/metabolism , HLA-G Antigens/pharmacology , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mitochondrial Trifunctional Protein, alpha Subunit/metabolism , Oxidoreductases/metabolism , Oxidoreductases/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Trophoblasts/metabolism
4.
Bioorg Med Chem ; 28(22): 115740, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33007553

ABSTRACT

Coenzyme A (CoA) is a highly selective inhibitor of the mitotic regulatory enzyme Aurora A kinase, with a novel mode of action. Herein we report the design and synthesis of analogues of CoA as inhibitors of Aurora A kinase. We have designed and synthesised modified CoA structures as potential inhibitors, combining dicarbonyl mimics of the pyrophosphate group with a conserved adenosine headgroup and different length pantetheine-based tail groups. An analogue with a -SH group at the end of the pantotheinate tail showed the best IC50, probably due to the formation of a covalent bond with Aurora A kinase Cys290.


Subject(s)
Aurora Kinase A/antagonists & inhibitors , Coenzyme A/pharmacology , Diphosphates/pharmacology , Drug Design , Pantetheine/pharmacology , Protein Kinase Inhibitors/pharmacology , Aurora Kinase A/metabolism , Coenzyme A/chemical synthesis , Coenzyme A/chemistry , Diphosphates/chemistry , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Pantetheine/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
5.
Bull Exp Biol Med ; 170(1): 58-63, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33237527

ABSTRACT

Mutations in pank2 gene encoding pantothenate kinase 2 determine a pantothenate kinase-associated neurodegeneration, a rare disorder characterized by iron deposition in the globus pallidus. To extend our previous work, we performed microinjections of a new pank2-specific morpholino to zebrafish embryos and thoroughly analyzed vasculature development. Vessels development was severely perturbed in the head, trunk, and tail, where blood accumulation was remarkable and associated with dilation of the posterior cardinal vein. This phenotype was specific as confirmed by p53 expression analysis and injection of the same morpholino in pank2-mutant embryos. We can conclude that pank2 gene is involved in vasculature development in zebrafish embryos. The comprehension of the underlining mechanisms could be of relevance for understanding of pantothenate kinase-associated neurodegeneration.


Subject(s)
Blood Vessels/metabolism , Coenzyme A/pharmacology , Globus Pallidus/metabolism , Pantothenate Kinase-Associated Neurodegeneration/prevention & control , Phosphotransferases (Alcohol Group Acceptor)/genetics , Animals , Blood Vessels/growth & development , Blood Vessels/pathology , Disease Models, Animal , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Globus Pallidus/blood supply , Globus Pallidus/drug effects , Globus Pallidus/pathology , Head/blood supply , Head/growth & development , Humans , Morpholinos/administration & dosage , Morpholinos/genetics , Morpholinos/metabolism , Pantothenate Kinase-Associated Neurodegeneration/genetics , Pantothenate Kinase-Associated Neurodegeneration/metabolism , Pantothenate Kinase-Associated Neurodegeneration/pathology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Tail/blood supply , Tail/growth & development , Tail/metabolism , Torso/blood supply , Torso/growth & development , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Zebrafish
6.
Nat Chem Biol ; 11(10): 784-92, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26322826

ABSTRACT

The metabolic cofactor coenzyme A (CoA) gained renewed attention because of its roles in neurodegeneration, protein acetylation, autophagy and signal transduction. The long-standing dogma is that eukaryotic cells obtain CoA exclusively via the uptake of extracellular precursors, especially vitamin B5, which is intracellularly converted through five conserved enzymatic reactions into CoA. This study demonstrates an alternative mechanism that allows cells and organisms to adjust intracellular CoA levels by using exogenous CoA. Here CoA was hydrolyzed extracellularly by ectonucleotide pyrophosphatases to 4'-phosphopantetheine, a biologically stable molecule able to translocate through membranes via passive diffusion. Inside the cell, 4'-phosphopantetheine was enzymatically converted back to CoA by the bifunctional enzyme CoA synthase. Phenotypes induced by intracellular CoA deprivation were reversed when exogenous CoA was provided. Our findings answer long-standing questions in fundamental cell biology and have major implications for the understanding of CoA-related diseases and therapies.


Subject(s)
Caenorhabditis elegans/metabolism , Coenzyme A/biosynthesis , Drosophila/metabolism , Pantetheine/analogs & derivatives , Animals , Caenorhabditis elegans/growth & development , Cell Line , Coenzyme A/blood , Coenzyme A/pharmacology , Coenzyme A Ligases/metabolism , Drosophila/cytology , Drosophila/growth & development , Female , HEK293 Cells , Humans , Longevity/physiology , Male , Mice, Inbred C57BL , Pantetheine/blood , Pantetheine/metabolism , Pantetheine/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism
7.
Exp Parasitol ; 134(3): 374-80, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23578856

ABSTRACT

Our previous observation that NADP-dependent secondary alcohol dehydrogenase (ADH-1) is down-regulated in metronidazole-resistant Trichomonas vaginalis isolates prompted us to further characterise the enzyme. In addition to its canonical enzyme activity as a secondary alcohol dehydrogenase, a pronounced, so far unknown, background NADPH-oxidising activity in absence of any added substrate was observed when the recombinant enzyme or T. vaginalis extract were used. This activity was strongly enhanced at low oxygen concentrations. Unexpectedly, all functions of ADH-1 were efficiently inhibited by coenzyme A which is a cofactor of a number of key enzymes in T. vaginalis metabolism, i.e. pyruvate:ferredoxin oxidoreductase (PFOR). These observations could be extended to Entamoeba histolytica and Tritrichomonas foetus, both of which have a homologue of ADH-1, but not to Giardia lamblia which lacks an NADP-dependent secondary alcohol dehydrogenase. Although we could not identify the substrate of the observed background activity, we propose that ADH-1 functions as a major sink for NADPH in microaerophilic parasites at low oxygen tension.


Subject(s)
Alcohol Oxidoreductases/metabolism , Entamoeba histolytica/enzymology , Trichomonas vaginalis/enzymology , Tritrichomonas foetus/enzymology , 2-Propanol/metabolism , 2-Propanol/pharmacology , Acetaldehyde/metabolism , Acetone/metabolism , Alcohol Oxidoreductases/antagonists & inhibitors , Alcohol Oxidoreductases/genetics , Coenzyme A/pharmacology , DNA, Protozoan/genetics , Entamoeba histolytica/genetics , Gene Expression Regulation, Enzymologic , Giardia lamblia/enzymology , Giardia lamblia/genetics , Hydrogen-Ion Concentration , Inhibitory Concentration 50 , Kinetics , NADP/metabolism , Oxidation-Reduction , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Trichomonas vaginalis/genetics , Tritrichomonas foetus/genetics
8.
Life Sci Alliance ; 6(12)2023 12.
Article in English | MEDLINE | ID: mdl-37833072

ABSTRACT

The tumor microenvironment is a dynamic network of stromal, cancer, and immune cells that interact and compete for resources. We have previously identified the Vanin1 pathway as a tumor suppressor of sarcoma development via vitamin B5 and coenzyme A regeneration. Using an aggressive sarcoma cell line that lacks Vnn1 expression, we showed that the administration of pantethine, a vitamin B5 precursor, attenuates tumor growth in immunocompetent but not nude mice. Pantethine boosts antitumor immunity, including the polarization of myeloid and dendritic cells towards enhanced IFNγ-driven antigen presentation pathways and improved the development of hypermetabolic effector CD8+ T cells endowed with potential antitumor activity. At later stages of treatment, the effect of pantethine was limited by the development of immune cell exhaustion. Nevertheless, its activity was comparable with that of anti-PD1 treatment in sensitive tumors. In humans, VNN1 expression correlates with improved survival and immune cell infiltration in soft-tissue sarcomas, but not in osteosarcomas. Pantethine could be a potential therapeutic immunoadjuvant for the development of antitumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes , Sarcoma , Humans , Mice , Animals , Coenzyme A/pharmacology , Pantothenic Acid/pharmacology , Sarcoma/drug therapy , Tumor Microenvironment
9.
Biochemistry ; 51(39): 7699-711, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-22954034

ABSTRACT

Disruption of the unusual thiol-based redox homeostasis mechanisms in Staphylococcus aureus represents a unique opportunity to identify new metabolic processes and new targets for intervention. Targeting uncommon aspects of CoASH biosynthetic and redox functions in S. aureus, the antibiotic CJ-15,801 has recently been demonstrated to be an antimetabolite of the CoASH biosynthetic pathway in this organism; CoAS-mimetics containing α,ß-unsaturated sulfone and carboxyl moieties have also been exploited as irreversible inhibitors of S. aureus coenzyme A-disulfide reductase (SaCoADR). In this work we have determined the crystal structures of three of these covalent SaCoADR-inhibitor complexes, prepared by inactivation of wild-type enzyme during turnover. The structures reveal the covalent linkage between the active-site Cys43-S(γ) and C(ß) of the vinyl sulfone or carboxyl moiety. The full occupancy of two inhibitor molecules per enzyme dimer, together with kinetic analyses of the wild-type/C43S heterodimer, indicates that half-sites-reactivity is not a factor during normal catalytic turnover. Further, we provide the structures of SaCoADR active-site mutants; in particular, Tyr419'-OH plays dramatic roles in directing intramolecular reduction of the Cys43-SSCoA redox center, in the redox asymmetry observed for the two FAD per dimer in NADPH titrations, and in catalysis. The two conformations observed for the Ser43 side chain in the C43S mutant structure lend support to a conformational switch for Cys43-S(γ) during its catalytic Cys43-SSCoA/Cys43-SH redox cycle. Finally, the structures of the three inhibitor complexes provide a framework for design of more effective inhibitors with therapeutic potential against several major bacterial pathogens.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Coenzyme A/chemistry , Coenzyme A/pharmacology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Staphylococcus aureus/enzymology , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Molecular Docking Simulation , Mutation , NADH, NADPH Oxidoreductases/chemistry , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , Oxidation-Reduction , Protein Multimerization , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcus aureus/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics
10.
Pharmacol Ther ; 238: 108282, 2022 10.
Article in English | MEDLINE | ID: mdl-36130624

ABSTRACT

Efferocytosis (clearance of apoptotic cells by phagocytosis without inducing inflammation and autoimmunity) is an important mechanism in the resolution of inflammatory processes. Efficient efferocytosis inhibits the accumulation of apoptotic cells/debris and maintains homeostasis before the onset of necrosis (secondary necrosis), which promotes inflammation or injury. Moreover, the detection and clearance of apoptotic cells can promote anti-inflammatory responses. Defective efferocytosis is involved in the pathogenesis of several diseases, such as atherosclerosis, chronic inflammation, autoimmunity and cancer. Statins are 3-hydroxy-3-methylglutaryl coenzyme A-reductase inhibitors which exert cholesterol-lowering effects plus multiple pleiotropic properties, such as inhibition of inflammation and macrophage proliferation. Statins exhibit anti-inflammatory properties by reducing both the prenylation of signaling molecules with downregulation of gene expression and the expression of adhesion molecules, as well as the levels of cytokines and chemokines. Additionally, statins suppress the prenylation of GTPases, such as Rac-1, as a positive regulator of efferocytosis, and RhoA, as a negative regulator of efferocytosis. However, statins alter the membrane balance of Rho GTPases in efferocytosis toward Rac-1. Efferocytosis has modifiable targets, which can be exploited for the treatment of several diseases, although limited attention has been given to the mechanisms by which statins regulate efferocytosis and the resulting therapeutic implications. In this review, we will elaborate on the mechanisms underlying the modulation of apoptotic cell clearance by statins, which, in turn, inhibits uncontrolled inflammation and ensuing diseases.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Apoptosis , Cholesterol , Coenzyme A/pharmacology , Cytokines , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Inflammation/drug therapy , Necrosis/drug therapy , Oxidoreductases , Phagocytosis/physiology , rho GTP-Binding Proteins
11.
Hum Exp Toxicol ; 41: 9603271221125934, 2022.
Article in English | MEDLINE | ID: mdl-36171180

ABSTRACT

The resistance of glioblastoma to chemotherapy remains a significant clinical problem. Targeting alternative pathways such as protein prenylation is known to be effective against many cancers. Fluvastatin is a potent competitive inhibitor of 3-hydroxy-3-methylglutaryl- CoA (HMG-CoA) reductase, thereby inhibits prenylation. We demonstrate that fluvastatin alone effectively inhibits proliferation and induces apoptosis in multiple human glioblastoma cell lines. The combination index analysis shows that fluvastatin acts synergistically with common chemotherapy drugs for glioblastoma: temozolomide and irinotecan. We further show that fluvastatin acts on glioblastoma through inhibiting prenylation-dependent Ras activation. The combination of fluvastatin and low dose temozolomide resulted in remarkable inhibition of glioblastoma tumor in mice throughout the whole treatment duration without causing toxicity. Such combinatorial effects provide the basis for utilizing these FDA-approved drugs as a potential clinical approach in overcoming resistance and improving glioblastoma treatment.


Subject(s)
Glioblastoma , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Animals , Coenzyme A/pharmacology , Coenzyme A/therapeutic use , Drug Resistance, Neoplasm , Fatty Acids, Monounsaturated/pharmacology , Fatty Acids, Monounsaturated/therapeutic use , Fluvastatin/pharmacology , Fluvastatin/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Irinotecan/pharmacology , Irinotecan/therapeutic use , Mice , Oxidoreductases , Protein Prenylation , Temozolomide/pharmacology
12.
Redox Biol ; 56: 102446, 2022 10.
Article in English | MEDLINE | ID: mdl-36057161

ABSTRACT

AIMS: Metabolic switching during heart development contributes to postnatal cardiomyocyte (CM) cell cycle exit and loss of regenerative capacity in the mammalian heart. Metabolic control has potential for developing effective CM proliferation strategies. We sought to determine whether lactate dehydrogenase A (LDHA) regulated CM proliferation by inducing metabolic reprogramming. METHODS AND RESULTS: LDHA expression was high in P1 hearts and significantly decreased during postnatal heart development. CM-specific LDHA knockout mice were generated using CRISPR/Cas9 technology. CM-specific LDHA knockout inhibited CM proliferation, leading to worse cardiac function and a lower survival rate in the neonatal apical resection model. In contrast, CM-specific overexpression of LDHA promoted CM proliferation and cardiac repair post-MI. The α-MHC-H2B-mCh/CAG-eGFP-anillin system was used to confirm the proliferative effect triggered by LDHA on P7 CMs and adult hearts. Metabolomics, proteomics and Co-IP experiments indicated that LDHA-mediated succinyl coenzyme A reduction inhibited succinylation-dependent ubiquitination of thioredoxin reductase 1 (Txnrd1), which alleviated ROS and thereby promoted CM proliferation. In addition, flow cytometry and western blotting showed that LDHA-driven lactate production created a beneficial cardiac regenerative microenvironment by inducing M2 macrophage polarization. CONCLUSIONS: LDHA-mediated metabolic reprogramming promoted CM proliferation by alleviating ROS and inducing M2 macrophage polarization, indicating that LDHA might be an effective target for promoting cardiac repair post-MI.


Subject(s)
Myocardial Infarction , Myocytes, Cardiac , Animals , Cell Proliferation , Coenzyme A/pharmacology , Lactate Dehydrogenase 5 , Lactates/metabolism , Lactates/pharmacology , Macrophages/metabolism , Mammals , Mice , Mice, Knockout , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Thioredoxin Reductase 1/metabolism
13.
Drug Des Devel Ther ; 16: 3071-3085, 2022.
Article in English | MEDLINE | ID: mdl-36118165

ABSTRACT

Background: Dihydromyricetin (DHM) exerts protective effects in various brain diseases. The aim of this research was to investigate the biological role of DHM in cerebral ischemia reperfusion (I/R) injury. Methods: We generated a rat model of cerebral I/R injury by performing middle cerebral artery occlusion/reperfusion (MCAO/R). The neurological score and brain water content of the experimental rats was then evaluated. The infarct volume and extent of apoptosis in brain tissues was then assessed by 2,3,5-triphenyltetrazolium (TTC) and TdT-mediated dUTP nick end labeling (TUNEL) staining. Hippocampal neuronal cells (HT22) were subjected to oxygen-glucose deprivation/reperfusion (OGD/R) and cell counting kit-8 (CCK-8) assays and flow cytometry were performed to detect cell viability and apoptosis. The levels of lipid reactive oxygen species (ROS) and iron were detected and the expression levels of key proteins were assessed by Western blotting. Results: DHM obviously reduced neurological deficits, brain water content, infarct volume and cell apoptosis in the brain tissues of MCAO/R rats. DHM repressed ferroptosis and inhibited the sphingosine kinase 1 (SPHK1)/mammalian target of rapamycin (mTOR) pathway in MCAO/R rats. In addition, DHM promoted cell viability and repressed apoptosis in OGD/R-treated HT22 cells. DHM also suppressed the levels of lipid ROS and intracellular iron in OGD/R-treated HT22 cells. The expression levels of glutathione peroxidase 4 (GPX4) was enhanced while the levels of acyl-CoA synthetase long-chain family member 4 (ACSL4) and phosphatidylethanolamine binding protein 1 (PEBP1) were reduced in OGD/R-treated HT22 cells in the presence of DHM. Moreover, the influence conferred by DHM was abrogated by the overexpression of SPHK1 or treatment with MHY1485 (an activator of mTOR). Conclusion: This research demonstrated that DHM repressed ferroptosis by inhibiting the SPHK1/mTOR signaling pathway, thereby alleviating cerebral I/R injury. Our findings suggest that DHM may be a candidate drug for cerebral I/R injury treatment.


Subject(s)
Ferroptosis , Reperfusion Injury , Animals , Coenzyme A/metabolism , Coenzyme A/pharmacology , Coenzyme A/therapeutic use , Flavonols , Glucose/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Iron , Ligases/metabolism , Ligases/pharmacology , Ligases/therapeutic use , Lipids/pharmacology , Mammals/metabolism , Oxygen/metabolism , Phosphatidylethanolamine Binding Protein/metabolism , Phosphatidylethanolamine Binding Protein/pharmacology , Phosphatidylethanolamine Binding Protein/therapeutic use , Phospholipid Hydroperoxide Glutathione Peroxidase , Phosphotransferases (Alcohol Group Acceptor) , Rats , Reactive Oxygen Species/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Water
14.
Nutr Res ; 106: 101-118, 2022 10.
Article in English | MEDLINE | ID: mdl-36183668

ABSTRACT

Previously, our group found that the dietary trace mineral element selenium and vitamin B6 (VitB6) alone was involved in lipid metabolism. However, the effects of selenium combined with VitB6 on hyperlipidemia and lipid metabolism have not been reported until now. We hypothesized that selenium and VitB6 cosupplementation would alleviate the hyperlipidemic and hepatic dysfunction and with minimum side effects in a Sprague-Dawley rat model of hyperlipidemia induced by a high-fat diet. Our results showed that selenium combined with VitB6 could improve dyslipidemia and displayed better in vivo hypocholesterolemic abilities at early intervention. Moreover, cosupplementation reduced atherogenic indexes (atherogenic index and atherogenic index of plasm) and the ratio of ApoB/ApoA1. The liver function index aspartate aminotransferase in serum was reduced, as was and total cholesterol, triacylglycerol, and low-density lipoprotein cholesterol in liver. The intervention also increased the levels of ApoA1 in serum and high-density lipoprotein cholesterol of liver. In addition, the combination of selenium and VitB6 decreased liver lipid deposition and alleviated steatosis, reduced adipocyte size of white adipose tissue, increased the activities of hepatic lipase and total lipase and the hepatic 3-hydroxy-3-methyl glutaryl coenzyme A reductase (HMGR) level, decreased the hepatic mRNA transcription of lipogenic and regulatory genes including Srebf1 and downstream fat synthesis-related enzymes (Acc and Fasn) and cholesterol synthesis speed limiting enzyme Hmgr, increased the mRNA abundance of Lcat and Cyp7a1, increased the protein expression of SIRT1 and PPARα, and up-regulated the protein expression of sterol regulatory element-binding protein 1c in the livers of hyperlipidemia rats. We first demonstrated that oral selenium and VitB6 cosupplementation exerted synergism in lowering blood and liver lipid profiles and antiatherosclerotic effects in hyperlipidemic rats by reducing endogenous cholesterol and lipid synthesis, enhancing the transport of cholesterol to hepatocytes and promoting fatty acid beta oxidation.


Subject(s)
Fatty Liver , Hyperlipidemias , Selenium , Trace Elements , Animals , Apolipoproteins B , Aspartate Aminotransferases/metabolism , Cholesterol/metabolism , Cholesterol, HDL , Cholesterol, LDL/metabolism , Coenzyme A/metabolism , Coenzyme A/pharmacology , Coenzyme A/therapeutic use , Diet, High-Fat/adverse effects , Fatty Acids/metabolism , Fatty Liver/metabolism , Hyperlipidemias/drug therapy , Lipase/metabolism , Lipase/pharmacology , Lipase/therapeutic use , Lipid Metabolism , Liver/metabolism , Oxidoreductases/metabolism , Oxidoreductases/pharmacology , Oxidoreductases/therapeutic use , PPAR alpha/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Selenium/pharmacology , Selenium/therapeutic use , Sirtuin 1/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Trace Elements/pharmacology , Trace Elements/therapeutic use , Triglycerides/metabolism , Vitamin B 6 , Vitamins/pharmacology
15.
Biochemistry ; 50(26): 5893-904, 2011 Jul 05.
Article in English | MEDLINE | ID: mdl-21627110

ABSTRACT

1,4-Dihydroxy-2-naphthoyl-coenzyme A (DHNA-CoA) synthase, or MenB, catalyzes an intramolecular Claisen condensation involving two oxyanion intermediates in the biosynthetic pathway of menaquinone, an essential respiration electron transporter in many microorganisms. Here we report the finding that the DHNA-CoA product and its analogues bind and inhibit the synthase from Escherichia coli with significant ultraviolet--visible spectral changes, which are similar to the changes induced by deprotonation of the free inhibitors in a basic solution. Dissection of the structure--affinity relationships of the inhibitors identifies the hydroxyl groups at positions 1 (C1-OH) and 4 (C4-OH) of DHNA-CoA or their equivalents as the dominant and minor sites, respectively, for the enzyme--ligand interaction that polarizes or deprotonates the bound ligands to cause the observed spectral changes. In the meantime, spectroscopic studies with active site mutants indicate that C4-OH of the enzyme-bound DHNA-CoA interacts with conserved polar residues Arg-91, Tyr-97, and Tyr-258 likely through a hydrogen bonding network that also includes Ser-161. In addition, site-directed mutation of the conserved Asp-163 to alanine causes a complete loss of the ligand binding ability of the protein, suggesting that the Asp-163 side chain is most likely hydrogen-bonded to C1-OH of DHNA-CoA to provide the dominant polarizing effect. Moreover, this mutation also completely eliminates the enzyme activity, strongly supporting the possibility that the Asp-163 side chain provides a strong stabilizing hydrogen bond to the tetrahedral oxyanion, which takes a position similar to that of C1-OH of the enzyme-bound DHNA-CoA and is the second high-energy intermediate in the intracellular Claisen condensation reaction. Interestingly, both Arg-91 and Tyr-97 are located in a disordered loop forming part of the active site of all available DHNA-CoA synthase structures. Their involvement in the interaction with the small molecule ligands suggests that the disordered loop is folded in interaction with the substrates or reaction intermediates, supporting an induced-fit catalytic mechanism for the enzyme.


Subject(s)
Aspartic Acid , Conserved Sequence , Oxo-Acid-Lyases/chemistry , Oxo-Acid-Lyases/metabolism , Oxygen/metabolism , Spectrum Analysis , Vitamin K 2/metabolism , Absorption , Bacteria/enzymology , Catalytic Domain , Coenzyme A/chemistry , Coenzyme A/metabolism , Coenzyme A/pharmacology , Enzyme Stability , Hydrogen Bonding , Models, Molecular , Mutagenesis, Site-Directed , Oxo-Acid-Lyases/antagonists & inhibitors , Oxo-Acid-Lyases/genetics , Oxygen/chemistry , Phenols/chemistry , Phenols/metabolism , Protons
16.
J Biol Chem ; 285(27): 20615-24, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20452977

ABSTRACT

In the aerobic metabolism of aromatic substrates, oxygenases use molecular oxygen to hydroxylate and finally cleave the aromatic ring. In the case of the common intermediate benzoate, the ring cleavage substrates are either catechol (in bacteria) or 3,4-dihydroxybenzoate (protocatechuate, mainly in fungi). We have shown before that many bacteria, e.g. Azoarcus evansii, the organism studied here, use a completely different mechanism. This elaborate pathway requires formation of benzoyl-CoA, followed by an oxygenase reaction and a nonoxygenolytic ring cleavage. Benzoyl-CoA transformation is catalyzed by the iron-containing benzoyl-CoA oxygenase (BoxB) in conjunction with an FAD and iron-sulfur centers containing reductase (BoxA), which donates electrons from NADPH. Here we show that benzoyl-CoA oxygenase actually does not form the 2,3-dihydrodiol of benzoyl-CoA, as formerly postulated, but the 2,3-epoxide. An enoyl-CoA hydratase (BoxC) uses two molecules of water to first hydrolytically open the ring of 2,3-epoxybenzoyl-CoA, which may proceed via its tautomeric seven-membered oxepin ring form. Then ring C2 is hydrolyzed off as formic acid, yielding 3,4-dehydroadipyl-CoA semialdehyde. The semialdehyde is oxidized by a NADP(+)-dependent aldehyde dehydrogenase (BoxD) to 3,4-dehydroadipyl-CoA. Final products of the pathway are formic acid, acetyl-CoA, and succinyl-CoA. This overlooked pathway occurs in 4-5% of all bacteria whose genomes have been sequenced and represents an elegant strategy to cope with the high resonance energy of aromatic substrates by forming a nonaromatic epoxide.


Subject(s)
Benzoates/metabolism , Coenzyme A/pharmacology , Epoxy Compounds/metabolism , Acyl Coenzyme A/metabolism , Aerobiosis , Azoarcus/drug effects , Azoarcus/metabolism , Bacterial Proteins/metabolism , Chromatography, High Pressure Liquid , Chromosomes, Bacterial/genetics , DNA Primers , Ditiocarb/pharmacology , Flavin-Adenine Dinucleotide/metabolism , Mass Spectrometry , NADP/metabolism , Oxygen/metabolism , Oxygen Isotopes
17.
J Biol Chem ; 285(16): 11870-9, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20133948

ABSTRACT

The Arabidopsis thaliana genome contains two genes encoding the mitochondrial NAD-malic enzyme (NAD-ME), NAD-ME1 (At2g13560) and NAD-ME2 (At4g00570). The characterization of recombinant NAD-ME1 and -2 indicated that both enzymes assemble as active homodimers; however, a heterodimeric enzyme (NAD-MEH) can also be detected by electrophoretic studies. To analyze the metabolic contribution of each enzymatic entity, NAD-MEH was obtained by a co-expression-based recombinant approach, and its kinetic and regulatory properties were analyzed. The three NAD-MEs show similar kinetic properties, although they differ in the regulation by several metabolic effectors. In this regard, whereas fumarate activates NAD-ME1 and CoA activates NAD-ME2, both compounds act synergistically on NAD-MEH activity. The characterization of two chimeric enzymes between NAD-ME1 and -2 allowed specific domains of the primary structure, which are involved in the differential allosteric regulation, to be identified. NAD-ME1 and -2 subunits showed a distinct pattern of accumulation in the separate components of the floral organ. In sepals, the NAD-ME1 subunit is present at a slightly higher proportion than the NAD-ME2 subunit, and thus, NAD-MEH and NAD-ME1 act in concert in this tissue. On the other hand, NAD-ME2 is the only isoform present in anthers. In view of the different properties of NAD-ME1, -2, and -H, we suggest that mitochondrial NAD-ME activity may be regulated by varying native association in vivo, rendering enzymatic entities with distinct allosteric regulation to fulfill specific roles. The presence of three different NAD-ME entities, which originate by alternative associations of two subunits, is suggested to be a novel phenomenon unique to plant mitochondria.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Malate Dehydrogenase/chemistry , Malate Dehydrogenase/metabolism , Allosteric Regulation , Amino Acid Sequence , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Base Sequence , Coenzyme A/pharmacology , DNA Primers/genetics , Enzyme Activation/drug effects , Fumarates/pharmacology , Genes, Plant , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Malate Dehydrogenase/genetics , Mitochondria/enzymology , Models, Biological , Molecular Sequence Data , Protein Multimerization , Protein Subunits , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Tissue Distribution
18.
Biochim Biophys Acta ; 1800(3): 385-91, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19931596

ABSTRACT

BACKGROUND: 5-Hydroxydecanoate (5-HD) inhibits preconditioning, and it is assumed to be a selective inhibitor of mitochondrial ATP-sensitive K(+) (mitoK(ATP)) channels. However, 5-HD is a substrate for mitochondrial outer membrane acyl-CoA synthetase, which catalyzes the reaction: 5HD + CoA + ATP --> 5-HD-CoA (5-hydroxydecanoyl-CoA) + AMP + pyrophosphate. We aimed to determine whether the reactants or principal product of this reaction modulate sarcolemmal K(ATP) (sarcK(ATP)) channel activity. METHODS: Single sarcK(ATP) channel currents were measured in inside-out patches excised from rat ventricular myocytes. In addition, sarcK(ATP) channel activity was recorded in whole-cell configuration or in giant inside-out patches excised from oocytes expressing Kir6.2/SUR2A. RESULTS: 5-HD inhibited (IC(50) approximately 30 microM) K(ATP) channel activity, albeit only in the presence of (non-inhibitory) concentrations of ATP. Similarly, when the inhibitory effect of 0.2 mM ATP was reversed by 1 microM oleoyl-CoA, subsequent application of 5-HD blocked channel activity, but no effect was seen in the absence of ATP. Furthermore, we found that 1 microM coenzyme A (CoA) inhibited sarcK(ATP) channels. Using giant inside-out patches, which are weakly sensitive to "contaminating" CoA, we found that Kir6.2/SUR2A channels were insensitive to 5-HD-CoA. In intact myocytes, 5-HD failed to reverse sarcK(ATP) channel activation by either metabolic inhibition or rilmakalim. GENERAL SIGNIFICANCE: SarcK(ATP) channels are inhibited by 5-HD (provided that ATP is present) and CoA but insensitive to 5-HD-CoA. 5-HD is equally potent at "directly" inhibiting sarcK(ATP) and mitoK(ATP) channels. However, in intact cells, 5-HD fails to inhibit sarcK(ATP) channels, suggesting that mitochondria are the preconditioning-relevant targets of 5-HD.


Subject(s)
Coenzyme A/pharmacology , Decanoic Acids/pharmacology , Hydroxy Acids/pharmacology , KATP Channels/physiology , Sarcolemma/physiology , Acetate-CoA Ligase/metabolism , Acetate-CoA Ligase/pharmacology , Animals , Heart Ventricles/drug effects , KATP Channels/antagonists & inhibitors , KATP Channels/drug effects , Mice , Muscle Cells/drug effects , Muscle Cells/physiology , Potassium Channels, Inwardly Rectifying/drug effects , Potassium Channels, Inwardly Rectifying/physiology , Rats
19.
Bioorg Chem ; 39(1): 42-7, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21111442

ABSTRACT

Histone and protein acetylation catalyzed by p300/CBP transcriptional coactivator regulates a variety of key biological pathways. This study investigates the proposed Theorell-Chance or "hit-and-run" catalytic mechanism of p300/CBP histone acetyltransferase (HAT) using bisubstrate analogs. A range of histone peptide tail peptide-CoA conjugates with different length linkers were synthesized and evaluated as inhibitors of p300 HAT. We show that longer linkers between the histone tail peptide and the CoA substrate moieties appear to allow for dual engagement of the two binding surfaces. Results with D1625R/D1628R double mutant p300 HAT further confirm the requirement for a negatively charged surface on the enzyme to interact with the histone tail.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , p300-CBP Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/metabolism , Acetylation , Amino Acid Sequence , Coenzyme A/chemistry , Coenzyme A/pharmacology , Humans , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , Peptides/pharmacology , p300-CBP Transcription Factors/chemistry
20.
Neurosci Res ; 170: 114-121, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33316301

ABSTRACT

Spinal cord injury (SCI) induces severe motor and sensory dysfunction. We previously showed the neuroprotective effects of COA-Cl, a novel synthesized adenosine analog, in a rat stroke model. In this study, we evaluated the neuroprotective effects of COA-Cl in acute phase of SCI. SCI was induced in rats at the T9 vertebra by using a drop device. Rats were divided into acute and subacute groups. A 5-day dose of 6 mg/kg COA-Cl in saline was given to the acute group immediately after SCI and the subacute group 4 days after SCI. Motor function assessed by Basso-Beattie-Bresnahan scoring and inclined plane test improved significantly in the acute group while the subacute group did not. Histological evaluation and TUNEL staining revealed that both the cavity volume and apoptosis were significantly decreased in the acute group compared with the subacute group. In addition, pERK/ERK was increased in the acute group 7 days after SCI. These results suggest that COA-Cl exerts neuroprotective effects via the ERK pathway when administered in the acute phase after SCI, resulting in the recovery of motor function. COA-Cl could be a novel therapeutic agent for the acute phase of SCI.


Subject(s)
Neuroprotective Agents , Spinal Cord Injuries , Animals , Apoptosis , Coenzyme A/pharmacology , Disease Models, Animal , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Recovery of Function , Spinal Cord , Spinal Cord Injuries/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL