Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 591
Filter
1.
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
2.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 53(5): 805-814, 2022 Sep.
Article in Chinese | 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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
J Med Chem ; 64(12): 8263-8271, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34110812

ABSTRACT

Protein N-terminal acetyltransferase D (NatD, NAA40) that specifically acetylates the alpha-N-terminus of histone H4 and H2A has been implicated in various diseases, but no inhibitor has been reported for this important enzyme. Based on the acetyl transfer mechanism of NatD, we designed and prepared a series of highly potent NatD bisubstrate inhibitors by covalently linking coenzyme A to different peptide substrates via an acetyl or propionyl spacer. The most potent bisubstrate inhibitor displayed an apparent Ki value of 1.0 nM. Biochemical studies indicated that bisubstrate inhibitors are competitive to the peptide substrate and noncompetitive to the cofactor, suggesting that NatD undergoes an ordered Bi-Bi mechanism. We also demonstrated that these inhibitors are highly specific toward NatD, displaying about 1000-fold selectivity over other closely related acetyltransferases. High-resolution crystal structures of NatD bound to two of these inhibitors revealed the molecular basis for their selectivity and inhibition mechanism, providing a rational path for future inhibitor development.


Subject(s)
Coenzyme A/pharmacology , Enzyme Inhibitors/pharmacology , N-Terminal Acetyltransferase D/antagonists & inhibitors , Peptides/pharmacology , Amino Acid Sequence , Cell Line, Tumor , Coenzyme A/chemical synthesis , Coenzyme A/metabolism , Crystallography, X-Ray , Drug Design , Enzyme Assays , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Humans , Kinetics , Molecular Structure , N-Terminal Acetyltransferase D/chemistry , N-Terminal Acetyltransferase D/metabolism , Peptides/chemical synthesis , Peptides/metabolism , Protein Binding , Structure-Activity Relationship
11.
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
12.
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
13.
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
14.
Redox Biol ; 28: 101318, 2020 01.
Article in English | MEDLINE | ID: mdl-31546169

ABSTRACT

Aurora A kinase is a master mitotic regulator whose functions are controlled by several regulatory interactions and post-translational modifications. It is frequently dysregulated in cancer, making Aurora A inhibition a very attractive antitumor target. However, recently uncovered links between Aurora A, cellular metabolism and redox regulation are not well understood. In this study, we report a novel mechanism of Aurora A regulation in the cellular response to oxidative stress through CoAlation. A combination of biochemical, biophysical, crystallographic and cell biology approaches revealed a new and, to our knowledge, unique mode of Aurora A inhibition by CoA, involving selective binding of the ADP moiety of CoA to the ATP binding pocket and covalent modification of Cys290 in the activation loop by the thiol group of the pantetheine tail. We provide evidence that covalent CoA modification (CoAlation) of Aurora A is specific, and that it can be induced by oxidative stress in human cells. Oxidising agents, such as diamide, hydrogen peroxide and menadione were found to induce Thr 288 phosphorylation and DTT-dependent dimerization of Aurora A. Moreover, microinjection of CoA into fertilized mouse embryos disrupts bipolar spindle formation and the alignment of chromosomes, consistent with Aurora A inhibition. Altogether, our data reveal CoA as a new, rather selective, inhibitor of Aurora A, which locks this kinase in an inactive state via a "dual anchor" mechanism of inhibition that might also operate in cellular response to oxidative stress. Finally and most importantly, we believe that these novel findings provide a new rationale for developing effective and irreversible inhibitors of Aurora A, and perhaps other protein kinases containing appropriately conserved Cys residues.


Subject(s)
Aurora Kinase A/chemistry , Aurora Kinase A/metabolism , Coenzyme A/administration & dosage , Animals , Coenzyme A/chemistry , Coenzyme A/pharmacology , Crystallography, X-Ray , HEK293 Cells , Hep G2 Cells , Humans , Mice , Models, Molecular , Oxidative Stress , Phosphorylation , Protein Conformation , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism
15.
Neuromolecular Med ; 21(2): 120-131, 2019 06.
Article in English | MEDLINE | ID: mdl-30141000

ABSTRACT

Pantothenate Kinase-Associated Neurodegeneration (PKAN) is a genetic and early-onset neurodegenerative disorder characterized by iron accumulation in the basal ganglia. It is due to mutations in Pantothenate Kinase 2 (PANK2), an enzyme that catalyzes the phosphorylation of vitamin B5, first and essential step in coenzyme A (CoA) biosynthesis. Most likely, an unbalance of the neuronal levels of this important cofactor represents the initial trigger of the neurodegenerative process, yet a complete understanding of the connection between PANK2 malfunctioning and neuronal death is lacking. Most PKAN patients carry mutations in both alleles and a loss of function mechanism is proposed to explain the pathology. When PANK2 mutants were analyzed for stability, dimerization capacity, and enzymatic activity in vitro, many of them showed properties like the wild-type form. To further explore this aspect, we overexpressed the wild-type protein, two mutant forms with reduced kinase activity and two retaining the catalytic activity in zebrafish embryos and analyzed the morpho-functional consequences. While the wild-type protein had no effects, all mutant proteins generated phenotypes that partially resembled those observed in pank2 and coasy morphants and were rescued by CoA and vitamin B5 supplementation. The overexpression of PANK2 mutant forms appears to be associated with perturbation in CoA availability, irrespective of their catalytic activity.


Subject(s)
Embryonic Development/physiology , Motor Activity/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Animals , Animals, Genetically Modified , Coenzyme A/biosynthesis , Coenzyme A/pharmacology , Embryo, Nonmammalian/physiology , Humans , Loss of Function Mutation , Mutation, Missense , Pantothenic Acid/biosynthesis , Pantothenic Acid/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/genetics , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , Recombinant Proteins/metabolism , Transgenes , Up-Regulation , Zebrafish/embryology , Zebrafish Proteins/metabolism
16.
Histol Histopathol ; 32(2): 171-176, 2017 02.
Article in English | MEDLINE | ID: mdl-27121633

ABSTRACT

The aim of this study was to use functional and morphological analyses to evaluate the protective effect of coenzyme A (CoA) in cisplatin-induced toxicity in outer hair cells (OHC). Three groups of 8 guinea pigs were used: control (group I), cisplatin-treated (group II) and cisplatin + CoA-treated (group III). In groups II and III, a single ototoxic dose of cisplatin (10 mg/kg) was injected intraperitoneally. Group III was co-treated with CoA (900 µg/kg per day for 7 consecutive days). Electrocochleography (ECoG) recordings were made before and after the 7-day treatment period in all groups. After ECoG on day 7, all animals were anesthetized and the cochleae were removed and fixed for ultrastructural analysis. Cell damage in OHC was observed with transmission electron microscopy. Cisplatin induced a significant increase in auditory thresholds (p<0.001) compared to group I (control). In contrast, group III (cisplatin + CoA) had significantly reduced thresholds (p<0.001) compared to the group treated with cisplatin alone (group II).We found no significant differences between the control group and animals co-treated with cisplatin and CoA. The electron microscopy findings in OHC were consistent with these results. Ultrastructural analysis of OHC in group II showed morphological indications of necrosis, i.e. cytoplasmic swelling and vacuolation, and mitochondrial swelling. In group III the cell morphology of OHC was preserved, with ultrastructural characteristics similar to the control group. In conclusion, co-treatment with cisplatin with CoA inhibited antineoplastic-induced cytotoxicity in OHC in a guinea pig model.


Subject(s)
Auditory Threshold/drug effects , Cisplatin/toxicity , Coenzyme A/pharmacology , Hair Cells, Auditory, Outer/drug effects , Protective Agents/pharmacology , Animals , Audiometry, Evoked Response , Auditory Threshold/physiology , Cochlea/drug effects , Cochlea/physiology , Cochlea/ultrastructure , Guinea Pigs , Hair Cells, Auditory, Outer/physiology , Hair Cells, Auditory, Outer/ultrastructure
17.
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
18.
J Clin Lipidol ; 9(5): 692-7, 2015.
Article in English | MEDLINE | ID: mdl-26350816

ABSTRACT

BACKGROUND: New, safer, and more effective agents to treat hyperlipidemia and thereby prevent cardiovascular events are under research. OBJECTIVE: To evaluate the lipid-lowering effects and safety of a natural hypolipidemic compound, coenzyme A (CoA) capsule, in Chinese patients with moderate dyslipidemia, compared with pantethine. METHODS: Overall, 216 subjects (124 males and 92 females; age, 18-75 years) with moderate dyslipidemia (triglyceride [TG], 2.3-6.5 mmol/L) were randomly divided into 2 groups administered CoA 400 U/d (n = 111) or pantethine 600 U/d (n = 105). Blood lipoproteins, liver and renal function, blood glucose, and complete blood count were measured at baseline and after 4- and 8-week treatment. RESULTS: TG reduction was 26.0% with CoA and 17.4% with pantethine after 4 weeks and 33.3% and 16.5% after 8 weeks; compared with baseline, the reduction was significant (P < .01) in both groups. The difference between the 2 groups was significant at both 4 weeks (P = .0413) and 8 weeks (P < .001). Compared with baseline, total cholesterol and non-high-density lipoprotein cholesterol (non-HDL-C) were reduced, whereas HDL-C was increased with CoA after 8 weeks (all P < .05). Compared with pantethine, total cholesterol (P = .026) and non-HDL-C (P = .005) were significantly reduced after 8 weeks of CoA treatment. There was no statistical difference in low-density lipoprotein cholesterol or HDL-C between the 2 groups (P > .05) and no difference in blood glucose, hepatic or renal function, myopathy, or gastrointestinal tract symptoms. CONCLUSIONS: CoA can improve TG and other lipoprotein parameters to a greater extent than pantethine in moderate dyslipidemia, with no obvious adverse effects.


Subject(s)
Coenzyme A/adverse effects , Coenzyme A/pharmacology , Hyperlipidemias/drug therapy , Pantetheine/analogs & derivatives , Safety , Adolescent , Adult , Aged , Coenzyme A/therapeutic use , Double-Blind Method , Female , Humans , Hyperlipidemias/blood , Lipoproteins/blood , Male , Middle Aged , Pantetheine/adverse effects , Pantetheine/pharmacology , Pantetheine/therapeutic use , Treatment Outcome , Triglycerides/blood , Young Adult
19.
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
20.
J Clin Endocrinol Metab ; 98(2): E275-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23293333

ABSTRACT

OBJECTIVES: The aim of the study was to evaluate the lipid-lowering effects and clinical safety of a natural hypolipidemic compound, coenzyme A (CoA) capsule, in Chinese patients with moderate dyslipidemia. METHODS: A total of 244 subjects (170 males and 74 females; aged 18-75 y) having moderate dyslipidemia (triglyceride [TG], 2.3-6.5 mmol · L(-1)) were randomly divided into 3 groups, to which placebo (group A, n = 81), CoA 200 U/d (group B, n = 79), and CoA 400 U/d (group C, n = 84) were administered, respectively. Blood lipoproteins, liver and renal functions, blood glucose, and complete blood count were measured at the baseline and after 4 or 8 weeks of treatment. RESULTS: After treatment for 4 weeks, TG was reduced by 5.1, 15.7, and 14.4% in groups A, B, and C, respectively. After treatment for 8 weeks, TG decreased .9, 21.7, and 36.1%, respectively. Compared with group A, the primary efficacy outcome TG in groups B and C significantly decreased (P < .01), and the difference between groups B and C was also significant (P < .01). Plasma total cholesterol, low-density lipoprotein cholesterol, and high-density lipoprotein cholesterol were not significantly different. Furthermore, there was no difference in blood glucose, hepatic and renal function test parameters, incidence of myopathy, or gastrointestinal tract symptoms among the 3 groups. CONCLUSION: CoA can effectively reduce plasma TG levels in subjects with moderate dyslipidemia and has no obvious adverse effect.


Subject(s)
Anticholesteremic Agents/therapeutic use , Coenzyme A/therapeutic use , Hyperlipidemias/drug therapy , Lipids/blood , Adolescent , Adult , Aged , Anticholesteremic Agents/pharmacology , Blood Glucose , Coenzyme A/pharmacology , Double-Blind Method , Female , Humans , Hyperlipidemias/blood , Hyperlipidemias/physiopathology , Kidney/drug effects , Kidney/physiopathology , Liver/drug effects , Liver/physiopathology , Male , Middle Aged , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...