Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 387
Filter
2.
Int J Mol Sci ; 25(3)2024 Feb 04.
Article in English | MEDLINE | ID: mdl-38339168

ABSTRACT

Differentiation-inducing factor 1 (DIF-1), found in Dictyostelium discoideum, has antiproliferative and glucose-uptake-promoting activities in mammalian cells. DIF-1 is a potential lead for the development of antitumor and/or antiobesity/antidiabetes drugs, but the mechanisms underlying its actions have not been fully elucidated. In this study, we searched for target molecules of DIF-1 that mediate the actions of DIF-1 in mammalian cells by identifying DIF-1-binding proteins in human cervical cancer HeLa cells and mouse 3T3-L1 fibroblast cells using affinity chromatography and liquid chromatography-tandem mass spectrometry and found mitochondrial malate dehydrogenase (MDH2) to be a DIF-1-binding protein in both cell lines. Since DIF-1 has been shown to directly inhibit MDH2 activity, we compared the effects of DIF-1 and the MDH2 inhibitor LW6 on the growth of HeLa and 3T3-L1 cells and on glucose uptake in confluent 3T3-L1 cells in vitro. In both HeLa and 3T3-L1 cells, DIF-1 at 10-40 µM dose-dependently suppressed growth, whereas LW6 at 20 µM, but not at 2-10 µM, significantly suppressed growth in these cells. In confluent 3T3-L1 cells, DIF-1 at 10-40 µM significantly promoted glucose uptake, with the strongest effect at 20 µM DIF-1, whereas LW6 at 2-20 µM significantly promoted glucose uptake, with the strongest effect at 10 µM LW6. Western blot analyses showed that LW6 (10 µM) and DIF-1 (20 µM) phosphorylated and, thus, activated AMP kinase in 3T3-L1 cells. Our results suggest that MDH2 inhibition can suppress cell growth and promote glucose uptake in the cells, but appears to promote glucose uptake more strongly than it suppresses cell growth. Thus, DIF-1 may promote glucose uptake, at least in part, via direct inhibition of MDH2 and a subsequent activation of AMP kinase in 3T3-L1 cells.


Subject(s)
Glucose , Malate Dehydrogenase , Animals , Humans , Mice , 3T3-L1 Cells/drug effects , 3T3-L1 Cells/metabolism , Adenylate Kinase/metabolism , Dictyostelium/metabolism , Glucose/metabolism , HeLa Cells/drug effects , HeLa Cells/metabolism , Malate Dehydrogenase/antagonists & inhibitors , Malate Dehydrogenase/metabolism , Mammals/metabolism
3.
Biomolecules ; 12(10)2022 10 03.
Article in English | MEDLINE | ID: mdl-36291624

ABSTRACT

Ischemia-reperfusion injury is the leading cause of acute kidney injury. Reactive oxygen species (ROS) production causes cell death or senescence. In cultures of primary human renal tubular epithelial cells (RPTECs) subjected to anoxia-reoxygenation, inhibition of the Krebs cycle at the level of malate dehydrogenase-2 (MDH-2) decreases hypoxia-inducible factor-1α and oxidative stress and protects from apoptotic or ferroptotic cell death. Inhibition of MDH-2 decreased reoxygenation-induced upregulation of p53 and p21, restored the levels of the proliferation marker Ki-67, and prevented the upregulation of the senescence marker beta-galactosidase and interleukin-1ß production. MDH-2 inhibition reduced the reoxygenation-induced upregulation of ATP, but the alterations of critical cell metabolism enzymes allowed enough ATP production to prevent cell energy collapse. Thus, inhibition of the Krebs cycle at the level of MDH-2 protects RPTECs from anoxia-reoxygenation-induced death or senescence. MDH-2 may be a promising pharmaceutical target against ischemia-reperfusion injury.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Malate Dehydrogenase , Reperfusion Injury , Humans , Adenosine Triphosphate/metabolism , Apoptosis , beta-Galactosidase/metabolism , Epithelial Cells/metabolism , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-1beta/metabolism , Ki-67 Antigen/metabolism , Malate Dehydrogenase/antagonists & inhibitors , Pharmaceutical Preparations/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Tumor Suppressor Protein p53/metabolism
4.
Bioorg Med Chem ; 50: 116458, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34687983

ABSTRACT

Parasitic diseases remain a major public health concern for humans, claiming millions of lives annually. Although different treatments are required for these diseases, drug usage is limited due to the development of resistance and toxicity, which necessitate alternative therapies. It has been shown in the literature that parasitic lactate dehydrogenases (LDH) and malate dehydrogenases (MDH) have unique pharmacological selective and specificity properties compared to other isoforms, thus highlighting them as viable therapeutic targets involved in aerobic and anaerobic glycolytic pathways. LDH and MDH are important therapeutic targets for invasive parasites because they play a critical role in the progression and development of parasitic diseases. Any strategy to impede these enzymes would be fatal to the parasites, paving the way to develop and discover novel antiparasitic agents. This review aims to highlight the importance of parasitic LDH and MDH as therapeutic drug targets in selected obligate apicoplast parasites. To the best of our knowledge, this review presents the first comprehensive review of LDH and MDH as potential antiparasitic targets for drug development studies.


Subject(s)
Antiparasitic Agents/pharmacology , Drug Development , L-Lactate Dehydrogenase/antagonists & inhibitors , Malate Dehydrogenase/antagonists & inhibitors , Animals , Antiparasitic Agents/chemical synthesis , Antiparasitic Agents/chemistry , Cryptosporidium parvum/drug effects , Cryptosporidium parvum/enzymology , Humans , L-Lactate Dehydrogenase/metabolism , Malate Dehydrogenase/metabolism , Molecular Structure , Parasitic Sensitivity Tests , Plasmodium/drug effects , Plasmodium/enzymology , Schistosoma/drug effects , Schistosoma/enzymology , Toxoplasma/drug effects , Toxoplasma/enzymology , Trichomonas vaginalis/drug effects , Trichomonas vaginalis/enzymology
5.
Bioorg Chem ; 115: 105258, 2021 10.
Article in English | MEDLINE | ID: mdl-34392176

ABSTRACT

Hsp90 (i.e., Heat shock protein 90) is a well-established therapeutic target for several diseases, ranging from misfolding-related disfunctions to cancer. In this framework, we have developed in recent years a family of benzofuran compounds that act as Hsp90 allosteric modulators. Such molecules can interfere with the stability of some relevant Hsp90 client oncoproteins, showing a low µM cytotoxic activity in vitro in cancer cell lines. Here we identify the target profile of these chemical probes by means of chemical proteomics, which established MDH2 (mitochondrial malate dehydrogenase) as an additional relevant cellular target that might help elucidate the molecular mechanism of their citotoxicity. Western blotting, DARTS (i.e., Drug Affinity Responsive Target Stability) and enzymatic assays data confirmed a dose-dependent interaction of MDH2 with several members of the benzofuran Hsp90 modulators family and a computational model allowed to interpret the observed interactions.


Subject(s)
Antineoplastic Agents/pharmacology , Benzofurans/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Malate Dehydrogenase/antagonists & inhibitors , Allosteric Regulation/drug effects , Antineoplastic Agents/chemistry , Benzofurans/chemistry , Dose-Response Relationship, Drug , HSP90 Heat-Shock Proteins/metabolism , Humans , Malate Dehydrogenase/metabolism , Models, Molecular , Molecular Structure , Structure-Activity Relationship
6.
Cell Metab ; 33(5): 1027-1041.e8, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33770508

ABSTRACT

Mitochondria have an independent genome (mtDNA) and protein synthesis machinery that coordinately activate for mitochondrial generation. Here, we report that the Krebs cycle intermediate fumarate links metabolism to mitobiogenesis through binding to malic enzyme 2 (ME2). Mechanistically, fumarate binds ME2 with two complementary consequences. First, promoting the formation of ME2 dimers, which activate deoxyuridine 5'-triphosphate nucleotidohydrolase (DUT). DUT fosters thymidine generation and an increase of mtDNA. Second, fumarate-induced ME2 dimers abrogate ME2 monomer binding to mitochondrial ribosome protein L45, freeing it for mitoribosome assembly and mtDNA-encoded protein production. Methylation of the ME2-fumarate binding site by protein arginine methyltransferase-1 inhibits fumarate signaling to constrain mitobiogenesis. Notably, acute myeloid leukemia is highly dependent on mitochondrial function and is sensitive to targeting of the fumarate-ME2 axis. Therefore, mitobiogenesis can be manipulated in normal and malignant cells through ME2, an unanticipated governor of mitochondrial biomass production that senses nutrient availability through fumarate.


Subject(s)
Fumarates/metabolism , Malate Dehydrogenase/metabolism , Mitochondria/metabolism , Animals , Cell Line , Citric Acid Cycle , DNA, Mitochondrial/metabolism , Dimerization , Humans , Leukemia/pathology , Leukemia/veterinary , Malate Dehydrogenase/antagonists & inhibitors , Malate Dehydrogenase/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mitochondria/genetics , Protein Binding , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Pyrophosphatases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Ribosomal Proteins/metabolism , Thymidine/metabolism
7.
J Med Chem ; 64(7): 4109-4116, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33761256

ABSTRACT

Small molecule colloidal aggregates adsorb and partially denature proteins, inhibiting them artifactually. Oddly, this inhibition is typically time-dependent. Two mechanisms might explain this: low concentrations of the colloid and enzyme might mean low encounter rates, or colloid-based protein denaturation might impose a kinetic barrier. These two mechanisms should have different concentration dependencies. Perplexingly, when enzyme concentration was increased, incubation times actually lengthened, inconsistent with both models and with classical chemical kinetics of solution species. We therefore considered molecular crowding, where colloids with lower protein surface density demand a shorter incubation time than more crowded colloids. To test this, we grew and shrank colloid surface area. As the surface area shrank, the incubation time lengthened, while as it increased, the converse was true. These observations support a crowding effect on protein binding to colloidal aggregates. Implications for drug delivery and for detecting aggregation-based inhibition will be discussed.


Subject(s)
Bacterial Proteins/metabolism , Colloids/metabolism , Malate Dehydrogenase/metabolism , beta-Lactamases/metabolism , Adsorption , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Colloids/chemistry , Enzyme Assays , Fulvestrant/chemistry , Kinetics , Malate Dehydrogenase/antagonists & inhibitors , Malate Dehydrogenase/chemistry , Protein Binding , Sorafenib/chemistry , beta-Lactamases/chemistry
8.
Bioorg Chem ; 110: 104779, 2021 05.
Article in English | MEDLINE | ID: mdl-33689977

ABSTRACT

Hypoxia inducible factor-1 (HIF-1) is a pivotal transcription factor, which is strongly correlated with the induction of angiogenesis, tumor survival, metastasis, and cell proliferation, making it a pivotal therapeutic target for solid tumor therapeutic agents. Herein, a new series of multi-functional chemical probes were designed including principal groups, viz. adamantyl and indene, at various locations of the parent compound LW6. Molecular docking studies were performed on the designed compounds and their relationship with HIF-1α and malate dehydrogenase 2 (MDH2). Inhibition of MDH2 by our compounds was expected to decrease the NADH level. Indeed, treatment of the breast cancer cell line 4T1 led to a strong reduction of the NADH concentration. The greatest reduction in NADH production in mitochondria was observed with (E)-3-(4-((3r, 5r, 7r)-adamantan-1-yl) phenoxy)-N-(5-(piperidine-1-carbonyl)-1, 4-dihydroindeno [1, 2-c] pyrazol-3-yl) acrylamide (18: IC50 = 59 nM), and has the best inhibitory potential under hypoxic conditions (MCF-7: IC50 = 57 nM). This compound also gave one of the highest docking "higher than the score obtained with LW6 in parallel (-31.63 kcal/mol) in the initial docking runs (PDB Code: 4WLO). Other related compounds with good yields were also synthesized from docking results, and all the synthesized compounds (14, 18, 22, 26, 29, 30) were evaluated in vitro on human adenocarcinoma cell lines.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Indenes/pharmacology , Malate Dehydrogenase/antagonists & inhibitors , Molecular Docking Simulation , Pyrazoles/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Indenes/chemical synthesis , Indenes/chemistry , Malate Dehydrogenase/metabolism , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Structure-Activity Relationship
9.
Int J Mol Sci ; 21(19)2020 Sep 28.
Article in English | MEDLINE | ID: mdl-32998265

ABSTRACT

Budding at the tumor invasive front has been correlated with the malignant properties of many cancers. Malic enzyme 1 (ME1) promotes the Warburg effect in cancer cells and induces epithelial-mesenchymal transition (EMT) in oral squamous cell carcinoma (OSCC). Therefore, we investigated the role of ME1 in tumor budding in OSCC. Tumor budding was measured in 96 human OSCCs by immunostaining for an epithelial marker (AE1/AE3), and its expression was compared with that of ME1. A significant correlation was observed between tumor budding and ME1 expression. The correlation increased with the progression of cancer. In human OSCC cells, lactate secretion decreased when lactate fermentation was suppressed by knockdown of ME1 and lactate dehydrogenase A or inhibition of pyruvate dehydrogenase (PDH) kinase. Furthermore, the extracellular pH increased, and the EMT phenotype was suppressed. In contrast, when oxidative phosphorylation was suppressed by PDH knockdown, lactate secretion increased, extracellular pH decreased, and the EMT phenotype was promoted. Induction of chemical hypoxia in OSCC cells by CoCl2 treatment resulted in increased ME1 expression along with HIF1α expression and promotion of the EMT phenotype. Hypoxic conditions also increased matrix metalloproteinases expression and decreased mitochondrial membrane potential, mitochondrial oxidative stress, and extracellular pH. Furthermore, the hypoxic treatment resulted in the activation of Yes-associated protein (YAP), which was abolished by ME1 knockdown. These findings suggest that cancer cells at the tumor front in hypoxic environments increase their lactate secretion by switching their energy metabolism from oxidative phosphorylation to glycolysis owing to ME1 overexpression, decrease in extracellular pH, and YAP activation. These alterations enhance EMT and the subsequent tumor budding. Tumor budding and ME1 expression are thus considered useful markers of OSCC malignancy, and ME1 is expected to be a relevant target for molecular therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinoma, Squamous Cell/genetics , Gene Expression Regulation, Neoplastic , Glycolysis/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Malate Dehydrogenase/genetics , Mouth Neoplasms/genetics , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing/metabolism , Aged , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Lymphatic Metastasis , Malate Dehydrogenase/antagonists & inhibitors , Malate Dehydrogenase/metabolism , Male , Middle Aged , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Oxidative Phosphorylation , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/antagonists & inhibitors , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Symporters/antagonists & inhibitors , Symporters/genetics , Symporters/metabolism , Transcription Factors/metabolism , YAP-Signaling Proteins
10.
FEBS Lett ; 594(10): 1631-1644, 2020 05.
Article in English | MEDLINE | ID: mdl-32232843

ABSTRACT

Radical S-adenosylmethionine (SAM) domain-containing protein 2 (RSAD2; viperin) is a key enzyme in innate immune responses that is highly expressed in response to viral infection and inflammatory stimuli in many cell types. Recently, it was found that RSAD2 catalyses transformation of cytidine triphosphate (CTP) to its analogue 3'-deoxy-3',4'-didehydro-CTP (ddhCTP). The cellular function of this metabolite is unknown. Here, we analysed the extra- and intracellular metabolite levels in human induced pluripotent stem cell (hiPSC)-derived macrophages using high-resolution LC-MS/MS. The results together with biochemical assays and molecular docking simulations revealed that ddhCTP inhibits the NAD+ -dependent activity of enzymes including that of the housekeeping enzyme glyceraldehyde 3-phosphate dehydrogenase (GAPDH). We propose that ddhCTP regulates cellular metabolism in response to inflammatory stimuli such as viral infection, pointing to a broader function of RSAD2 than previously thought.


Subject(s)
Cytidine Triphosphate/metabolism , Macrophages/enzymology , NAD/metabolism , Proteins/metabolism , Adenosine Diphosphate/metabolism , Binding Sites , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/antagonists & inhibitors , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/metabolism , Macrophages/cytology , Macrophages/metabolism , Malate Dehydrogenase/antagonists & inhibitors , Malate Dehydrogenase/metabolism , Models, Molecular , Oxidoreductases Acting on CH-CH Group Donors
11.
World J Microbiol Biotechnol ; 36(2): 24, 2020 Jan 21.
Article in English | MEDLINE | ID: mdl-31965331

ABSTRACT

The study evaluated the antibacterial activity of chlorogenic acid (CA) against Salmonella Enteritidis S1, a foodborne pathogen in chilled fresh chicken. Its minimum inhibitory concentration for S. Enteritidis S1 was 2 mM. 1 MIC CA treatment reduced the viable count of S. Enteritidis S1 by 3 log cfu/g in chilled fresh chicken. Scanning electron microscopy examination indicated that CA induced the cell envelope damage of S. Enteritidis S1. Following this, 1-N-Phenylnaphthylamine assay and LPS content analysis indicated that CA induced the permeability of outer membrane (OM). Confocal laser scanning microscopy examination further demonstrated that CA acted on the inner membrane (IM). To support this, the release of intracellular protein and ATP after CA treatment was also observed. CA also suppressed the activities of malate dehydrogenase and succinate dehydrogenase, two main metabolic enzymes in TCA cycle and electron transport chain. Thus, damage of intracelluar and outer membranes as well as disruption of cell metabolism resulted in cell death eventually. The finding suggested that CA has the potential to be developed as a preservative to control S. Enteritidis associated foodborne diseases.


Subject(s)
Anti-Bacterial Agents/pharmacology , Chlorogenic Acid/pharmacology , Salmonella enteritidis/drug effects , Animals , Bacterial Proteins/antagonists & inhibitors , Cell Membrane/drug effects , Chickens/microbiology , Colony Count, Microbial , Gene Expression Regulation, Bacterial/drug effects , Malate Dehydrogenase/antagonists & inhibitors , Microbial Sensitivity Tests , Microbial Viability/drug effects , Microscopy, Electron, Scanning , Salmonella enteritidis/enzymology , Salmonella enteritidis/growth & development , Succinate Dehydrogenase/antagonists & inhibitors
12.
Physiol Plant ; 168(2): 278-288, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31152557

ABSTRACT

NADPH is an essential cofactor in many physiological processes. Fruit ripening is caused by multiple biochemical pathways in which, reactive oxygen and nitrogen species (ROS/RNS) metabolism is involved. Previous studies have demonstrated the differential modulation of nitric oxide (NO) and hydrogen sulfide (H2 S) content during sweet pepper (Capsicum annuum L.) fruit ripening, both of which regulate NADP-isocitrate dehydrogenase activity. To gain a deeper understanding of the potential functions of other NADPH-generating components, we analyzed glucose-6-phosphate dehydrogenase (G6PDH) and 6-phosphogluconate dehydrogenase (6PGDH), which are involved in the oxidative phase of the pentose phosphate pathway (OxPPP) and NADP-malic enzyme (NADP-ME). During fruit ripening, G6PDH activity diminished by 38%, while 6PGDH and NADP-ME activity increased 1.5- and 2.6-fold, respectively. To better understand the potential regulation of these NADP-dehydrogenases by H2 S, we obtained a 50-75% ammonium-sulfate-enriched protein fraction containing these proteins. With the aid of in vitro assays, in the presence of H2 S, we observed that, while NADP-ME activity was inhibited by up to 29-32% using 2 and 5 mM Na2 S as H2 S donor, G6PDH and 6PGDH activities were unaffected. On the other hand, NO donors, S-nitrosocyteine (CysNO) and DETA NONOate also inhibited NADP-ME activity by 35%. These findings suggest that both NADP-ME and 6PGDH play an important role in maintaining the supply of NADPH during pepper fruit ripening and that H2 S and NO partially modulate the NADPH-generating system.


Subject(s)
Capsicum/enzymology , Hydrogen Sulfide/pharmacology , Malate Dehydrogenase/antagonists & inhibitors , NADP , Nitric Oxide/pharmacology , Capsicum/drug effects , Fruit/drug effects , Fruit/enzymology , Glucosephosphate Dehydrogenase , Phosphogluconate Dehydrogenase , Plant Proteins/antagonists & inhibitors
13.
Small ; 15(27): e1900860, 2019 07.
Article in English | MEDLINE | ID: mdl-31111667

ABSTRACT

Widely used silver nanoparticles (AgNPs) are readily accessible to biological fluids and then surrounded by proteins. However, interactions between AgNPs and proteins are poorly understood. Two dehydrogenases, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and malate dehydrogenase (MDH), are chosen to investigate these interactions. Ag bound to thiol groups of these enzymes significantly decreases the number of free thiols available. Dose-dependent inhibition of enzyme activities is observed in both AgNPs and Ag+ treatments. Based on the concentration required to inhibit 50% activity, GAPDH and MDH are 24-30 fold more sensitive to Ag+ than to AgNPs suggesting that the measured 4.2% Ag+ containing AgNPs can be responsible for the enzymes inhibition. GAPDH, with a thiol group in its active site, is more sensitive to Ag than MDH, displaying many thiol groups but none in its active site, suggesting that thiol groups at the active site strongly determines the sensitivity of enzymes toward AgNPs. In contrast, the dramatic changes of circular dichroism spectra show that the global secondary structure of MDH under AgNPs treatment is more altered than that of GAPDH. In summary, this study shows that the thiol groups and their location on these dehydrogenases are crucial for the AgNPs effects.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Malate Dehydrogenase/metabolism , Metal Nanoparticles/chemistry , Silver/chemistry , Sulfhydryl Compounds/chemistry , Animals , Dithiothreitol/pharmacology , Dynamic Light Scattering , Glyceraldehyde-3-Phosphate Dehydrogenases/antagonists & inhibitors , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Hydrodynamics , Malate Dehydrogenase/antagonists & inhibitors , Malate Dehydrogenase/chemistry , Mass Spectrometry , Metal Nanoparticles/ultrastructure , Models, Molecular , Particle Size , Protein Structure, Secondary , Rabbits , Silver/pharmacology , Static Electricity , Substrate Specificity/drug effects , Swine
14.
Cancer Res ; 79(8): 1884-1898, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30765601

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is driven by metabolic changes in pancreatic cells caused by oncogenic mutations and dysregulation of p53. PDAC cell lines and PDAC-derived xenografts grow as a result of altered metabolic pathways, changes in stroma, and autophagy. Selective targeting and inhibition of one of these may open avenues for the development of new therapeutic strategies. In this study, we performed a genome-wide siRNA screen in a PDAC cell line using endogenous autophagy as a readout and identified several regulators of autophagy that were required for autophagy-dependent PDAC cell survival. Validation of two promising candidates, MPP7 (MAGUK p55 subfamily member 7, a scaffolding protein involved in cell-cell contacts) and MDH1 (cytosolic Malate dehydrogenase 1), revealed their role in early stages of autophagy during autophagosome formation. MPP7 was involved in the activation of YAP1 (a transcriptional coactivator in the Hippo pathway), which in turn promoted autophagy, whereas MDH1 was required for maintenance of the levels of the essential autophagy initiator serine-threonine kinase ULK1, and increased in the activity upon induction of autophagy. Our results provide a possible explanation for how autophagy is regulated by MPP7 and MDH1, which adds to our understanding of autophagy regulation in PDAC. SIGNIFICANCE: This study identifies and characterizes MPP7 and MDH1 as novel regulators of autophagy, which is thought to be responsible for pancreatic cancer cell survival.


Subject(s)
Autophagy , Carcinoma, Pancreatic Ductal/pathology , Gene Expression Regulation, Neoplastic , Malate Dehydrogenase/metabolism , Membrane Proteins/metabolism , Pancreatic Neoplasms/pathology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Proliferation , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Malate Dehydrogenase/antagonists & inhibitors , Malate Dehydrogenase/genetics , Membrane Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , RNA, Small Interfering/genetics , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , YAP-Signaling Proteins
15.
Sci Rep ; 8(1): 14268, 2018 09 24.
Article in English | MEDLINE | ID: mdl-30250042

ABSTRACT

Cytosolic Malic Enzyme (ME1) provides reduced NADP for anabolism and maintenance of redox status. To examine the role of ME1 in tumor genesis of the gastrointestinal tract, we crossed mice having augmented intestinal epithelial expression of ME1 (ME1-Tg mice) with ApcMin/+ mice to obtain male ApcMin/+/ME1-Tg mice. ME1 protein levels were significantly greater within gut epithelium and adenomas of male ApcMin/+/ME1-Tg than ApcMin/+ mice. Male ApcMin/+/ME1-Tg mice had larger and greater numbers of adenomas in the small intestine (jejunum and ileum) than male ApcMin/+ mice. Male ApcMin/+/ME1-Tg mice exhibited greater small intestine crypt depth and villus length in non-adenoma regions, correspondent with increased KLF9 protein abundance in crypts and lamina propria. Small intestines of male ApcMin/+/ME1-Tg mice also had enhanced levels of Sp5 mRNA, suggesting Wnt/ß-catenin pathway activation. A small molecule inhibitor of ME1 suppressed growth of human CRC cells in vitro, but had little effect on normal rat intestinal epithelial cells. Targeting of ME1 may add to the armentarium of therapies for cancers of the gastrointestinal tract.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Carcinogenesis/genetics , Colonic Neoplasms/genetics , Gastrointestinal Tract/metabolism , Malate Dehydrogenase/genetics , Animals , Cell Proliferation/genetics , Colonic Neoplasms/pathology , DNA-Binding Proteins/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gastrointestinal Tract/pathology , Gene Expression Regulation, Neoplastic/genetics , Humans , Intestinal Mucosa , Malate Dehydrogenase/antagonists & inhibitors , Mice , Oncogenes , Rats , Transcription Factors/genetics
16.
IUBMB Life ; 70(11): 1076-1083, 2018 11.
Article in English | MEDLINE | ID: mdl-30160039

ABSTRACT

Reprogrammed metabolic profile is a biochemical fingerprint of cancerous cells, which represents one of the "hallmarks of cancer." The aberrant expression pattern of enzymatic machineries orchestrates metabolic activities into a platform that ultimately promotes cellular growth, survival, and proliferation. The NADP(+)-dependent mitochondrial malic enzyme 2 (ME2) has been widely appreciated due to its function as a provider of pyruvate and reducing power to the cell for biosynthesis of fatty acids and nucleotides along with maintenance of redox balance. Multiple lines of evidences have indicated that ME2 is a bonafide therapeutic target and novel biomarker which plays critical role during tumorigenesis. The objective of this review is to provide an update on the cancer-specific role of ME2 in order to explore its potential for therapeutic opportunities. Furthermore, we have discussed the potential of genetic and pharmacological inhibitors of ME2 in the light of previous research work for therapeutic advancements in cancer treatment. It is contemplated that additional investigations should focus on the use of ME2 inhibitors in combinational therapies as rational combinations of metabolic inhibitors and chemotherapy may have the ability to cure cancer. © 2018 IUBMB Life, 70(11):1076-1083, 2018.


Subject(s)
Antineoplastic Agents/therapeutic use , Malate Dehydrogenase/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/enzymology , Humans , Neoplasms/pathology , Prognosis
17.
FEMS Yeast Res ; 18(7)2018 11 01.
Article in English | MEDLINE | ID: mdl-30052989

ABSTRACT

An oenological strain of Saccharomyces cerevisiae was previously shown to produce a 5-10 kDa peptidic fraction responsible for the inhibition of malolactic fermentation (MLF). In the present study, we aim to further purify the anti-MLF peptides of this fraction. The yeast fermented synthetic grape juice medium was fractionated by ammonium sulfate precipitation combined with ultrafiltration. The 5-10 kDa fraction recovered at a saturation degree of 60%-80% was the only fraction that inhibited both the bacterial growth and the malate consumption in vivo. It also inhibited the malolactic enzyme activity in vitro at a pH range between 3.5 and 6.7. Therefore, it was purified by both anion and cation exchange chromatography. The eluates that inhibited the malolactic enzyme activity in vitro were migrated on Tricine SDS-PAGE and the protein bands were excised and sequenced by LC-MS/MS. The sequencing revealed nine peptides originating from eight proteins of S. cerevisiae. Two GAPDH cationic fragments of 0.9 and 1.373 kDa having a pI of 10.5 and 11 respectively, Wtm2p and Utr2p anionic fragments of 2.42 kDa with a pI of 3.5 and 4 respectively were thought to contribute the most to the MLF inhibition.


Subject(s)
Fermentation , Malate Dehydrogenase/antagonists & inhibitors , Malates/metabolism , Peptides/chemistry , Saccharomyces cerevisiae/chemistry , Amino Acid Sequence , Fermentation/drug effects , Fungal Proteins/chemistry , Hydrogen-Ion Concentration , Lactic Acid/biosynthesis , Molecular Weight , Oenococcus/drug effects , Oenococcus/growth & development , Oenococcus/metabolism , Peptides/pharmacology , Vitis/metabolism
18.
PLoS One ; 13(4): e0195011, 2018.
Article in English | MEDLINE | ID: mdl-29694407

ABSTRACT

Malaria remains a major threat to human health, as strains resistant to current therapeutics are discovered. Efforts in finding new drug targets are hampered by the lack of sufficiently specific tools to provide target validation prior to initiating expensive drug discovery projects. Thus, new approaches that can rapidly enable drug target validation are of significant interest. In this manuscript we present the crystal structure of malate dehydrogenase from Plasmodium falciparum (PfMDH) at 2.4 Å resolution and structure-based mutagenic experiments interfering with the inter-oligomeric interactions of the enzyme. We report decreased thermal stability, significantly decreased specific activity and kinetic parameters of PfMDH mutants upon mutagenic disruption of either oligomeric interface. In contrast, stabilization of one of the interfaces resulted in increased thermal stability, increased substrate/cofactor affinity and hyperactivity of the enzyme towards malate production at sub-millimolar substrate concentrations. Furthermore, the presented data show that our designed PfMDH mutant could be used as specific inhibitor of the wild type PfMDH activity, as mutated PfMDH copies were shown to be able to self-incorporate into the native assembly upon introduction in vitro, yielding deactivated mutant:wild-type species. These data provide an insight into the role of oligomeric assembly in regulation of PfMDH activity and reveal that recombinant mutants could be used as probe tool for specific modification of the wild type PfMDH activity, thus offering the potential to validate its druggability in vivo without recourse to complex genetics or initial tool compounds. Such tool compounds often lack specificity between host or pathogen proteins (or are toxic in in vivo trials) and result in difficulties in assessing cause and effect-particularly in cases when the enzymes of interest possess close homologs within the human host. Furthermore, our oligomeric interference approach could be used in the future in order to assess druggability of other challenging human pathogen drug targets.


Subject(s)
Antimalarials/chemistry , Drug Discovery , Malate Dehydrogenase/chemistry , Plasmodium falciparum/enzymology , Amino Acid Sequence , Antimalarials/pharmacology , Binding Sites , Conserved Sequence , Gene Expression , Humans , Malate Dehydrogenase/antagonists & inhibitors , Malate Dehydrogenase/genetics , Models, Molecular , Molecular Conformation , Mutation , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Protein Binding , Recombinant Proteins , Substrate Specificity
19.
Int J Oncol ; 52(6): 1923-1933, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29620192

ABSTRACT

The present study investigated the possible tumor-suppressing function of microRNA (miR)-612 and the underlying molecular mechanism of its action in bladder cancer in vitro and in vivo. Reverse transcription­quantitative polymerase chain reaction (RT­qPCR) was carried out to quantify the expression levels of miR­612 in bladder cancer tissues and cell lines. The data demonstrated that the level of miR­612 expression was significantly reduced in bladder cancer tissues and cell lines, as compared with that in non­cancerous tissues and cells. Reduced miR­612 expression was associated with advanced tumor, lymph node and metastasis stages, and with distant metastasis of bladder cancer. A functional study revealed that transfection of cells with an miR­612 mimic suppressed bladder cancer cell growth, colony formation, migration, invasion and epithelial-mesenchymal transition. Bioinformatics analysis identified that miR­612 targeted the expression of malic enzyme 1 (ME1), and this was confirmed by western blot and luciferase reporter assay results. Furthermore, the ME1 expression levels were inversely associated with miR­612 expression in bladder cancer tissue specimens. In addition, knockdown of ME1 expression using ME1 siRNA mimicked the effect of ectopic miR­612 overexpression in bladder cancer cells in terms of tumor cell growth, migration and invasion. By contrast, ME1 overexpression weakened the inhibitory effect of the miR­612 mimic in bladder cancer cells. In conclusion, the present study demonstrated that miR­612 may function as a tumor suppressor in bladder cancer by targeting ME1 expression.


Subject(s)
Down-Regulation , Malate Dehydrogenase/genetics , Malate Dehydrogenase/metabolism , MicroRNAs/genetics , Urinary Bladder Neoplasms/pathology , 3' Untranslated Regions , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Malate Dehydrogenase/antagonists & inhibitors , Male , Mice , Neoplasm Staging , Neoplasm Transplantation , RNA, Small Interfering/pharmacology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
20.
Cancer Sci ; 109(6): 2036-2045, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29601126

ABSTRACT

Malic enzyme 1 (ME1) is a multifunctional protein involved in glycolysis, the citric acid cycle, NADPH production, glutamine metabolism, and lipogenesis. It is overexpressed in various cancers. We examined the expression of ME1 in 119 oral squamous cell carcinomas (OSCCs) using immunohistochemistry. Malic enzyme 1 expression was moderate to strong in 57 (48%) OSCCs and correlated with pT, pN, clinical stage, and histological grade. In 37 cases with prognostic evaluation, moderate to strong ME1 expression indicated a worse prognosis than did weak ME1 expression. Malic enzyme 1 knockdown or inactivation by lanthanide inhibited cell proliferation and motility and suppressed the epithelial-mesenchymal transition in HSC3 human OSCC cells. Knockdown of ME1 also shifted energy metabolism from aerobic glycolysis and lactate fermentation to mitochondrial oxidative phosphorylation, and the redox status from reductive to oxidative. In a mouse tumor model, lanthanide suppressed tumor growth and increased survival time. These findings reveal that ME1 is a valid target for molecular therapy in OSCC.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Cytosol/enzymology , Malate Dehydrogenase/biosynthesis , Mouth Neoplasms/enzymology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Disease Progression , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Lanthanoid Series Elements/pharmacology , Malate Dehydrogenase/antagonists & inhibitors , Malate Dehydrogenase/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Oligonucleotides, Antisense/genetics , Oxidation-Reduction/drug effects , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...