Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 930
Filter
1.
Molecules ; 29(9)2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38731521

ABSTRACT

Lactate dehydrogenase A (LDHA) primarily catalyzes the conversion between lactic acid and pyruvate, serving as a key enzyme in the aerobic glycolysis pathway of sugar in tumor cells. LDHA plays a crucial role in the occurrence, development, progression, invasion, metastasis, angiogenesis, and immune escape of tumors. Consequently, LDHA not only serves as a biomarker for tumor diagnosis and prognosis but also represents an ideal target for tumor therapy. Although LDHA inhibitors show great therapeutic potential, their development has proven to be challenging. In the development of LDHA inhibitors, the key active sites of LDHA are emphasized. Nevertheless, there is a relative lack of research on the amino acid residues around the active center of LDHA. Therefore, in this study, we investigated the amino acid residues around the active center of LDHA. Through structure comparison analysis, five key amino acid residues (Ala30, Met41, Lys131, Gln233, and Ala259) were identified. Subsequently, the effects of these five residues on the enzymatic properties of LDHA were investigated using site-directed mutagenesis. The results revealed that the catalytic activities of the five mutants varied to different degrees in both the reaction from lactic acid to pyruvate and pyruvate to lactic acid. Notably, the catalytic activities of LDHAM41G and LDHAK131I were improved, particularly in the case of LDHAK131I. The results of the molecular dynamics analysis of LDHAK131I explained the reasons for this phenomenon. Additionally, the optimum temperature of LDHAM41G and LDHAQ233M increased from 35 °C to 40 °C, whereas in the reverse reaction, the optimum temperature of LDHAM41G and LDHAK131I decreased from 70 °C to 60 °C. These findings indicate that Ala30, Met41, Lys131, Gln233, and Ala259 exert diverse effects on the catalytic activity and optimum temperature of LHDA. Therefore, these amino acid residues, in addition to the key catalytic site of the active center, play a crucial role. Considering these residues in the design and screening of LDHA inhibitors may lead to the development of more effective inhibitors.


Subject(s)
Catalytic Domain , Enzyme Inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Amino Acids/chemistry , Amino Acids/metabolism , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/metabolism , L-Lactate Dehydrogenase/chemistry , Lactate Dehydrogenase 5/metabolism , Lactate Dehydrogenase 5/antagonists & inhibitors , Lactate Dehydrogenase 5/chemistry , Pyruvic Acid/metabolism , Pyruvic Acid/chemistry , Mutagenesis, Site-Directed , Molecular Dynamics Simulation
2.
Science ; 377(6614): 1519-1529, 2022 09 30.
Article in English | MEDLINE | ID: mdl-36173860

ABSTRACT

Gain-of-function mutations in isocitrate dehydrogenase (IDH) in human cancers result in the production of d-2-hydroxyglutarate (d-2HG), an oncometabolite that promotes tumorigenesis through epigenetic alterations. The cancer cell-intrinsic effects of d-2HG are well understood, but its tumor cell-nonautonomous roles remain poorly explored. We compared the oncometabolite d-2HG with its enantiomer, l-2HG, and found that tumor-derived d-2HG was taken up by CD8+ T cells and altered their metabolism and antitumor functions in an acute and reversible fashion. We identified the glycolytic enzyme lactate dehydrogenase (LDH) as a molecular target of d-2HG. d-2HG and inhibition of LDH drive a metabolic program and immune CD8+ T cell signature marked by decreased cytotoxicity and impaired interferon-γ signaling that was recapitulated in clinical samples from human patients with IDH1 mutant gliomas.


Subject(s)
CD8-Positive T-Lymphocytes , Carcinogenesis , Glutarates , Isocitrate Dehydrogenase , Neoplasms , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Gain of Function Mutation , Glutarates/metabolism , Humans , Interferon-gamma/metabolism , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/metabolism , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism
3.
Blood ; 139(19): 2958-2971, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35176139

ABSTRACT

Translation is essential for megakaryocyte (MK) maturation and platelet production. However, how the translational pathways are regulated in this process remains unknown. In this study, we found that MK/platelet-specific lactate dehydrogenase A (LdhA) knockout mice exhibited an increased number of platelets with remarkably accelerated MK maturation and proplatelet formation. Interestingly, the role of LDHA in MK maturation and platelet formation did not depend on lactate content, which was the major product of LDHA. Mechanism studies revealed that LDHA interacted with eukaryotic elongation factor 2 (eEF2) in the cytoplasm, controlling the participation of eEF2 in translation at the ribosome. Furthermore, the interaction of LDHA and eEF2 was dependent on nicotinamide adenine dinucleotide (NADH), a coenzyme of LDHA. NADH-competitive inhibitors of LDHA could release eEF2 from the LDHA pool, upregulate translation, and enhance MK maturation in vitro. Among LDHA inhibitors, stiripentol significantly promoted the production of platelets in vivo under a physiological state and in the immune thrombocytopenia model. Moreover, stiripentol could promote platelet production from human cord blood mononuclear cell-derived MKs and also have a superposed effect with romiplostim. In short, this study shows a novel nonclassical function of LDHA in translation that may serve as a potential target for thrombocytopenia therapy.


Subject(s)
Elongation Factor 2 Kinase , L-Lactate Dehydrogenase , Megakaryocytes , Thrombocytopenia , Thrombopoiesis , Animals , Blood Platelets/cytology , Blood Platelets/metabolism , Elongation Factor 2 Kinase/blood , Elongation Factor 2 Kinase/metabolism , Enzyme Inhibitors/pharmacology , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/blood , L-Lactate Dehydrogenase/metabolism , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Mice, Knockout , NAD/metabolism , Peptide Elongation Factor 2/metabolism , Thrombocytopenia/blood , Thrombocytopenia/drug therapy , Thrombocytopenia/enzymology , Thrombocytopenia/metabolism , Thrombopoiesis/physiology
4.
Biomed Pharmacother ; 147: 112686, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35124385

ABSTRACT

Cancer is one of the main causes of human mortality and brain tumors, including invasive pituitary adenomas, medulloblastomas and glioblastomas are common brain malignancies with poor prognosis. Therefore, the development of innovative management strategies for refractory cancers and brain tumors is important. In states of mitochondrial dysfunction - commonly encountered in malignant cells - cells mostly shift to anaerobic glycolysis by increasing the expression of LDHA (Lactate Dehydrogenase-A) gene. Oxamate, an isosteric form of pyruvate, blocks LDHA activity by competing with pyruvate. By blocking LDHA, it inhibits protumorigenic cascades and also induces ROS (reactive oxygen species)-induced mitochondrial apoptosis of cancer cells. In preclinical studies, oxamate blocked the growth of invasive pituitary adenomas, medulloblastomas and glioblastomas. Oxamate also increases temozolomide and radiotherapy sensitivity of glioblastomas. Oxamate is highly polar, which may preclude its clinical utilization due to low penetrance through cell membranes. However, this obstacle could be overcome with nanoliposomes. Moreover, different oxamate analogs were developed which inhibit LDHC4, an enzyme also involved in cancer progression and germ cell physiology. Lastly, phenformin, an antidiabetic agent, exerts anticancer effects via complex I inhibition in the mitochondria and leading the overproduction of ROS. Oxamate combination with phenformin reduces the lactic acidosis-causing side effect of phenformin while inducing synergistic anticancer efficacy. In sum, oxamate as a single agent and more efficiently with phenformin has high potential to slow the progression of aggressive cancers with special emphasis to brain tumors.


Subject(s)
Brain Neoplasms/pathology , L-Lactate Dehydrogenase/antagonists & inhibitors , Oxamic Acid/pharmacology , Animals , Cell Line, Tumor , Glycolysis/physiology , Humans , L-Lactate Dehydrogenase/metabolism , Mitochondria/metabolism , Neoplasms/pathology , Phenformin/pharmacology , Radiation Tolerance/drug effects , Reactive Oxygen Species/metabolism , Temozolomide/pharmacology
5.
Bioorg Med Chem Lett ; 59: 128576, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35065235

ABSTRACT

Structure-based design was utilized to optimize 6,6-diaryl substituted dihydropyrone and hydroxylactam to obtain inhibitors of lactate dehydrogenase (LDH) with low nanomolar biochemical and single-digit micromolar cellular potencies. Surprisingly the replacement of a phenyl with a pyridyl moiety in the chemical structure revealed a new binding mode for the inhibitors with subtle conformational change of the LDHA active site. This led to the identification of a potent, cell-active hydroxylactam inhibitor exhibiting an in vivo pharmacokinetic profile suitable for mouse tumor xenograft study.


Subject(s)
Enzyme Inhibitors/pharmacology , L-Lactate Dehydrogenase/antagonists & inhibitors , Lactams/pharmacology , Animals , Cell Line , Dogs , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Humans , L-Lactate Dehydrogenase/metabolism , Lactams/chemistry , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Structure , Structure-Activity Relationship
6.
Immunology ; 165(2): 158-170, 2022 02.
Article in English | MEDLINE | ID: mdl-34606637

ABSTRACT

Treatment of myocarditis is often limited to symptomatic treatment due to unknown pathomechanisms. In order to identify new therapeutic approaches, the contribution of locked nucleic acid antisense oligonucleotides (LNA ASOs) in autoimmune myocarditis was investigated. Hence, A/J mice were immunized with cardiac troponin I (TnI) to induce experimental autoimmune myocarditis (EAM) and treated with LNA ASOs. The results showed an unexpected anti-inflammatory effect for one administered LNA ASO MB_1114 by reducing cardiac inflammation and fibrosis. The target sequence of MB_1114 was identified as lactate dehydrogenase B (mLDHB). For further analysis, mice received mLdhb-specific GapmeR during induction of EAM. Here, mice receiving the mLdhb-specific GapmeR showed increased protein levels of cardiac mLDHB and a reduced cardiac inflammation and fibrosis. The effect of increased cardiac mLDHB protein level was associated with a downregulation of genes of reactive oxygen species (ROS)-associated proteins, indicating a reduction in ROS. Here, the suppression of murine pro-apoptotic Bcl-2-associated X protein (mBax) was also observed. In our study, an unexpected anti-inflammatory effect of LNA ASO MB_1114 and mLdhb-specific GapmeR during induction of EAM could be demonstrated in vivo. This effect was associated with increased protein levels of cardiac mLDHB, mBax suppression and reduced ROS activation. Thus, LDHB and LNA ASOs may be considered as a promising target for directed therapy of myocarditis. Nevertheless, further investigations are necessary to clarify the mechanism of action of anti-inflammatory LDHB-triggered effects.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , L-Lactate Dehydrogenase/antagonists & inhibitors , Myocarditis/etiology , Myocarditis/metabolism , Oligonucleotides/pharmacology , Animals , Autoimmune Diseases/diagnosis , Autoimmune Diseases/drug therapy , Biomarkers , Biopsy , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Enzyme Inhibitors/pharmacology , Female , Immunohistochemistry , Inflammation Mediators/metabolism , Isoenzymes/antagonists & inhibitors , Mice , Myocarditis/diagnosis , Myocarditis/drug therapy , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacology , Reactive Oxygen Species/metabolism
7.
J Biomol Struct Dyn ; 40(11): 5159-5174, 2022 07.
Article in English | MEDLINE | ID: mdl-33416018

ABSTRACT

Malaria infection caused by Plasmodium falciparum is majorly responsible for millions of deaths in humans every year. Moreover, a rapid increase in resistance to existing drugs has posed an urgent need for new anti-malarials. Herein, we report the highly potent anti-malarial activity of benzopyrano(4,3-b)benzopyran derivatives, inspired from naturally occurring dependensin against chloroquine (CQ) sensitive and resistant P. falciparum strains. Chemically synthesized, four dependensin analogs 85(A-D) exhibited growth inhibition at nanomolar concentrations ranging from 63.96 to 725.8 nM by blocking the parasite development at the ring and early trophozoite stages. The growth inhibitory activity of dependensin analogs was correlated with their anti-plasmodial lactate dehydrogenase activity by computational analysis. Molecular docking, 50 ns simulation and a 2D-Quantitative Structure-Activity Relationship (2D-QSAR) modelling revealed the interaction with their putative target P. falciparum lactate dehydrogenase (PfLDH). Here, developing the predictive 2D descriptors such as thermodynamic, spatial, electronic, and topological with multiple linear regression analysis (MLRA), the structural requirements for potent and selective PfLDH inhibitory activity has been identified. The strong binding of compound 85D to the catalytic Nicotinamide adenine dinucleotide (NADH) binding pocket of the PfLDH further supported the PfLDH targeting potential of dependensin analogs. Overall, this study revealed a highly potent anti-malarial activity of benzopyrano(4,3-b)benzopyran derivatives with their putative anti-PfLDH activity.Communicated by Ramaswamy H. Sarma.


Subject(s)
Antimalarials , Benzopyrans , L-Lactate Dehydrogenase , Plasmodium falciparum , Antimalarials/chemistry , Antimalarials/pharmacology , Benzopyrans/pharmacology , L-Lactate Dehydrogenase/antagonists & inhibitors , Molecular Docking Simulation , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology
8.
Sci Rep ; 11(1): 21353, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34725423

ABSTRACT

Lactate dehydrogenase (LDH) catalyses the conversion of pyruvate to lactate and NADH to NAD+; it has two isoforms, LDHA and LDHB. LDHA is a promising target for cancer therapy, whereas LDHB is necessary for basal autophagy and cancer cell proliferation in oxidative and glycolytic cancer cells. To the best of our knowledge, selective inhibitors for LDHB have not yet been reported. Here, we developed a high-throughput mass spectrometry screening system using an LDHB enzyme assay by detecting NADH and NAD+. As a result, we identified a small-molecule LDHB selective inhibitor AXKO-0046, an indole derivative. This compound exhibited uncompetitive LDHB inhibition (EC50 = 42 nM). X-ray crystallography revealed that AXKO-0046 bound to the potential allosteric site away from the LDHB catalytic active site, suggesting that targeting the tetramerisation interface of the two dimers is critical for the enzymatic activity. AXKO-0046 and its derivatives can be used to validate LDHB-associated pathways in cancer metabolism.


Subject(s)
Enzyme Inhibitors/pharmacology , Indoles/pharmacology , L-Lactate Dehydrogenase/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Crystallography, X-Ray , Drug Discovery , Enzyme Inhibitors/chemistry , Humans , Indoles/chemistry , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/metabolism , L-Lactate Dehydrogenase/chemistry , L-Lactate Dehydrogenase/metabolism , Models, Molecular , Small Molecule Libraries/chemistry
9.
Chem Biol Interact ; 350: 109688, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34627786

ABSTRACT

Malaria remains one of the most important parasitic diseases in the world. The multidrug-resistant Plasmodium strains make the treatment currently available for malaria less effective. Therefore, the development of new drugs is necessary to overcome therapy resistance. Triazole derivatives exhibit several biological activities and provide a moiety that is promising from the biological perspective. Due to the structural similarity to NADH, it is believed that triazoles can bind to the active site of the Plasmodium lactate dehydrogenase (pLDH) enzyme. The present work evaluates the antimalarial activity of 1,2,3-triazole derivatives by in silico, in vitro, and in vivo studies. Preliminary in silico ADMET studies of the compounds demonstrated good pharmacokinetic properties. In silico docking analysis against LDH of Plasmodium berghei (PbLDH) showed that all compounds presented interactions with the catalytic residue in the active site and affinity similar to that presented by chloroquine; the most common antimalarial drug. Cytotoxicity and hemolysis by these derivatives were evaluated in vitro. The compounds 1, 2, 5, 8, and 9 proved to be non-cytotoxic in the performed tests. In vivo antimalarial activity was evaluated using mice infected with Plasmodium berghei NK65. The five compounds tested exhibited antimalarial activity until nine days post-infection. The compound 5 showed promising activities, with about 70% parasitemia suppression. Considering the in vitro and in vivo studies, we believe the compound 5 to be the most promising molecule for further studies in antimalarial chemotherapy.


Subject(s)
Antimalarials/chemical synthesis , Antimalarials/pharmacokinetics , Triazoles/chemical synthesis , Triazoles/pharmacokinetics , Animals , Antimalarials/toxicity , Catalytic Domain , Computer Simulation , Drug Evaluation, Preclinical , Female , Hemolysis/drug effects , Humans , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/chemistry , Macrophages, Peritoneal/drug effects , Malaria/drug therapy , Malaria/parasitology , Mice , Molecular Docking Simulation , Plasmodium berghei/drug effects , Plasmodium berghei/enzymology , Protein Structure, Quaternary , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Structure-Activity Relationship , Triazoles/toxicity
10.
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
11.
Exp Oncol ; 43(3): 270-273, 2021 09.
Article in English | MEDLINE | ID: mdl-34591427

ABSTRACT

BACKGROUND: Inhibition of aerobic glycolysis of cancer cells is considered a promising therapeutic strategy for the treatment of neoplasms. Some inhibitors of energy metabolism can affect not only tumor cells but also the functional polarization of tumor-associated macrophages, which may either enhance the antitumor effect of such agents or impair their antitumor efficacy. AIM: To investigate the effect of oxamate, a lactate dehydrogenase (LDH) inhibitor, on the polarization of peritoneal macrophages (PMP) in both intact mice and mice with transplanted Lewis lung carcinoma (LLC). MATERIALS AND METHODS: The low-metastatic LLC variant, LLC/R9, was transplanted to female C57Bl/6 mice. Sodium oxamate was used as the test agent at concentrations of 0.02, 0.2, and 2 mg/ml. Macrophage polarization in tumor-bearing mice was estimated on day 23 after tumor transplantation by assessing nitric oxide (NO) production and arginase activity as functional indices of PMPs polarization. RESULTS: Oxamate can affect the functional polarization of PMPs in both intact mice and animals with transplanted LLC/R9. Oxamate in all studied concentrations changed the markers of PMPs polarization in intact mice (decreasing NO levels and activating arginase activity) that indicated the stimulation of M2 polarization. In tumor-bearing animals, stimulation of M2 polarization is observed at low concentrations of oxamate (0.02 mg/ml), but its high concentrations (2.0 mg/ml) causes M1 polarization, which is characterized by three-fold increase in the level of NO and a decrease in the level of arginase activity. CONCLUSION: Oxamate, an inhibitor of LDH, can stimulate M2 polarization of peritoneal macrophages of mice bearing LLC in a dose-dependent manner.


Subject(s)
Carcinoma, Lewis Lung/immunology , L-Lactate Dehydrogenase/antagonists & inhibitors , Macrophage Activation/immunology , Macrophages, Peritoneal/immunology , Oxamic Acid/pharmacology , Animals , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Energy Metabolism , Female , Macrophage Activation/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Nitric Oxide/metabolism
12.
Cell Cycle ; 20(18): 1812-1827, 2021 09.
Article in English | MEDLINE | ID: mdl-34382917

ABSTRACT

The hypoxia-induced transcription factor HIF1 inhibits cell growth in normoxia through poorly understood mechanisms. A constitutive upregulation of hypoxia response is associated with increased malignancy, indicating a loss of antiproliferative effects of HIF1 in cancer cells. To understand these differences, we examined the control of cell cycle in primary human cells with activated hypoxia response in normoxia. Activated HIF1 caused a global slowdown of cell cycle progression through G1, S and G2 phases leading to the loss of mitotic cells. Cell cycle inhibition required a prolonged HIF1 activation and was not associated with upregulation of p53 or the CDK inhibitors p16, p21 or p27. Growth inhibition by HIF1 was independent of its Asn803 hydroxylation or the presence of HIF2. Antiproliferative effects of hypoxia response were alleviated by inhibition of lactate dehydrogenase and, more effectively, by boosting cellular production of NAD+, which was decreased by HIF1 activation. In comparison to normal cells, various cancer lines showed several fold-higher expressions of NAMPT, which is a rate-limiting enzyme in the main biosynthetic pathway for NAD+. Inhibition of NAMPT activity in overexpressor cancer cells sensitized them to antigrowth effects of HIF1. Thus, metabolic changes in cancer cells, such as enhanced NAD+ production, create resistance to growth-inhibitory activity of HIF1 permitting manifestation of its tumor-promoting properties.Abbreviations: DMOG: dimethyloxalylglycine, DM-NOFD: dimethyl N-oxalyl-D-phenylalanine, NMN: ß-nicotinamide mononucleotide.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Hypoxia/genetics , Cell Proliferation/genetics , Fibroblasts/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/metabolism , NAD/metabolism , Signal Transduction/genetics , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/pathology , Amino Acids, Dicarboxylic/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Hypoxia/drug effects , Cell Proliferation/drug effects , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Gene Knockdown Techniques/methods , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/metabolism , Lung Neoplasms/pathology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/metabolism , Signal Transduction/drug effects , Transfection
13.
Drug Discov Today ; 26(11): 2508-2514, 2021 11.
Article in English | MEDLINE | ID: mdl-34325010

ABSTRACT

Targeted-therapy failure in treating nonsmall cell lung cancer (NSCLC) frequently occurs because of the emergence of drug resistance and genetic mutations. The same mutations also result in aerobic glycolysis, which further antagonizes outcomes by localized increases in lactate, an immune suppressor. Recent evidence indicates that enzymatic lowering of lactate can promote an oncolytic immune microenvironment within the tumour. Here, we review factors relating to lactate expression in NSCLC and the utility of lactate oxidase (LOX) for governing therapeutic delivery, its role in lactate oxidation and turnover, and relationships between lactate depletion and immune cell populations. The lactate-rich characteristic of NSCLC provides an exploitable property to potentially improve NSCLC outcomes and design new therapeutic strategies to integrate with conventional therapies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lactic Acid/metabolism , Lung Neoplasms/metabolism , Tumor Microenvironment , Anaplastic Lymphoma Kinase/genetics , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Citric Acid Cycle , Drug Resistance, Neoplasm , Genes, erbB-1/genetics , Glucose/metabolism , Humans , Immune Checkpoint Inhibitors/therapeutic use , L-Lactate Dehydrogenase/antagonists & inhibitors , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Metabolic Networks and Pathways , Mixed Function Oxygenases/therapeutic use , Molecular Targeted Therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics
14.
Clin Transl Med ; 11(6): e467, 2021 06.
Article in English | MEDLINE | ID: mdl-34185423

ABSTRACT

BACKGROUND: Pancreatic adenocarcinoma (PAAD) a highly lethal malignancy. The current use of clinical parameters may not accurately predict the clinical outcome, which further renders the unsatisfactory therapeutic outcome. METHODS: In this study, we retrospectively analyzed the clinical-pathological characteristics and prognosis of 253 PAAD patients. Univariate, multivariate, and Kaplan-Meier survival analyses were conducted to assess risk factors and clinical outcomes. For functional study, we performed bidirectional genetic manipulation of lactate dehydrogenase A (LDHA) in PAAD cell lines to measure PAAD progression by both in vitro and in vivo assays. RESULTS: LDHA is particularly overexpressed in PAAD tissues and elevated serum LDHA-transcribed isoenzymes-5 (LDH-5) was associated with poorer patients' clinical outcomes. Genetic overexpression of LDHA promoted the proliferation and invasion in vitro, and tumor growth and metastasis in vivo in murine PAAD orthotopic models, while knockdown of LDHA exhibited opposite effects. LDHA-induced L-lactate production was responsible for the LDHA-facilitated PAAD progression. Mechanistically, LDHA overexpression reduced the phosphorylation of metabolic regulator AMPK and promoted the downstream mTOR phosphorylation in PAAD cells. Inhibition of mTOR repressed the LDHA-induced proliferation and invasion. A natural product berberine was selected as functional inhibitor of LDHA, which reduced activity and expression of the protein in PAAD cells. Berberine inhibited PAAD cells proliferation and invasion in vitro, and suppressed tumor progression in vivo. The restoration of LDHA attenuated the suppressive effect of berberine on PAAD. CONCLUSIONS: Our findings suggest that LDHA may be a novel biomarker and potential therapeutic target of human PAAD.


Subject(s)
Adenocarcinoma/drug therapy , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , L-Lactate Dehydrogenase/antagonists & inhibitors , Liver Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Animals , Apoptosis , Cell Movement , Cell Proliferation , Female , Glycolysis , Humans , Isoenzymes , Liver Neoplasms/enzymology , Liver Neoplasms/secondary , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Prognosis , Small Molecule Libraries/pharmacology , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Molecules ; 26(11)2021 05 23.
Article in English | MEDLINE | ID: mdl-34071141

ABSTRACT

The pathogenesis of colorectal cancer is a multifactorial process. Dysbiosis and the overexpression of COX-2 and LDHA are important effectors in the initiation and development of the disease through chromosomal instability, PGE2 biosynthesis, and induction of the Warburg effect, respectively. Herein, we report the in vitro testing of some new quinoxalinone and quinazolinone Schiff's bases as: antibacterial, COX-2 and LDHA inhibitors, and anticolorectal agents on HCT-116 and LoVo cells. Moreover, molecular docking and SAR analyses were performed to identify the structural features contributing to the biological activities. Among the synthesized molecules, the most active cytotoxic agent, (6d) was also a COX-2 inhibitor. In silico ADMET studies predicted that (6d) would have high Caco-2 permeability, and %HIA (99.58%), with low BBB permeability, zero hepatotoxicity, and zero risk of sudden cardiac arrest, or mutagenicity. Further, (6d) is not a potential P-gp substrate, instead, it is a possible P-gpI and II inhibitor, therefore, it can prevent or reverse the multidrug resistance of the anticancer drugs. Collectively, (6d) can be considered as a promising lead suitable for further optimization to develop anti-CRC agents or glycoproteins inhibitors.


Subject(s)
Colorectal Neoplasms/drug therapy , Quinazolinones/pharmacology , Quinoxalines/pharmacology , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Caco-2 Cells , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Drug Design , Drug Resistance, Multiple/drug effects , Humans , L-Lactate Dehydrogenase/antagonists & inhibitors , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship
16.
Acta Histochem ; 123(5): 151739, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34107386

ABSTRACT

Resveratrol is a kind of iPolyphenols widely existing in herbal medicine. Here we aim to investigate whether resveratrol can reduce the degree of myocardial ischemia/reperfusion (IR) injury and inhibit the development of oxidative stress, and elucidate the molecular mechanism of resveratrol in protecting myocardial cells. The primary rat cardiomyocytes were used to establish an ischemia/reperfusion model in vitro, and a series of routine biochemical experiments were conducted to explore the antioxidant and anti-apoptotic effects of resveratrol in myocardial ischemia-reperfusion injury. Compared with that of the simulated ischemia-refusion (SIR) group, cell viability in the SIR and resveratrol co-treatment groups increased significantly (P < 0.001), the release of lactate dehydrogenase (LDH) and creatine kinase MB (CKMB) decreased, the positive rate of reactive oxygen species (ROS) in cardiomyocytes decreased, and the concentration of catalase and glutathione peroxidase increased significantly (P < 0.001). Besides, resveratrol can activate PI3K/AKT signaling pathway. PI3K siRNA can inhibit the PI3K/AKT signaling mediated by resveratrol. The addition of resveratrol can significantly increase the activity of mitochondrial superoxide dismutase (SOD) and reduce the malondialdehyde (MDA), which indicates that the oxidative damage of mitochondria induced by resveratrol was significantly weakened. The mitochondrial functional changes induced by resveratrol can be reversed by PI3K siRNA. In conclusion, our study shows that resveratrol can reduce ROS in cardiomyocytes by PI3K/AKT signaling pathway activation, and effectively inhibit the apoptosis of cardiomyocytes, thus having a direct protective effect on cardiomyocytes under SR.


Subject(s)
Apoptosis , Mitochondria/metabolism , Myocardium/pathology , Phosphatidylinositol 3-Kinases/metabolism , Polyphenols/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Reperfusion Injury/pathology , Animals , Animals, Newborn , Cell Survival , Creatine Kinase, MB Form/biosynthesis , L-Lactate Dehydrogenase/antagonists & inhibitors , Male , Malondialdehyde/chemistry , Myocytes, Cardiac/cytology , RNA, Small Interfering/metabolism , Rats , Reactive Oxygen Species , Resveratrol/pharmacology , Signal Transduction
17.
J Ethnopharmacol ; 278: 114322, 2021 Oct 05.
Article in English | MEDLINE | ID: mdl-34118343

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Duan Teng Yimu decoction is a Chinese herbal medicine compound with proven therapeutic effects on inflammasome-related diseases, such as rheumatoid arthritis. This decoction consists of three Chinese herbal medicines, including Leonurus japonicus (L. japonicus), which promotes the blood circulation and exhibits detumescence activity, traditionally curing gynecologic and inflammasome diseases. AIM OF THE STUDY: To explore the anti-inflammasome activity and the underlying mechanisms of action of the compounds from L. japonicus. MATERIALS AND METHODS: A series of compounds were isolated from L. japonicus. Their anti-inflammasome activities were evaluated in macrophages that were co-stimulated by lipopolysaccharide (LPS) and NLRP3 inflammasome inducers. NLRP3 inflammasome formation and apoptosis speck like containing a CARD (ASC) oligomerization were evaluated by immunofluorescent microscopy and Western blot analysis. The regulation of autophagy after treatment of this compound was also evaluated. Lastly, in vivo activity of Leojaponin was analyzed in a mouse acute gouty arthritis model. RESULTS: Here we show that Leojaponin, a diterpenoid compound from L. japonicus, suppressed lactate dehydrogenase and IL-1ß release in Nigericin-stimulated macrophages in a pyroptosis model. Leojaponin inhibits NLRP3 inflammasome activation in both J774A.1 cells and bone marrow-derived macrophages in a dose dependent manner. Moreover, Leojaponin suppressed NLRP3-mediated ASC specks formation and ASC oligomerization. These activities of Leojaponin depend on restoration of autophagy via promoting RAPTOR phosphorylation. Furthermore, Leojaponin ameliorated monosodium urate (MSU)-induced acute gouty arthritis in vivo. CONCLUSION: Our findings suggest that Leojaponin inhibits NLRP3 inflammasome activation through enhancing autophagy via RAPTOR phosphorylation, thereby highlighting Leojaponin as a potent drug for inflammasome-related diseases.


Subject(s)
Arthritis, Gouty/drug therapy , Autophagy/drug effects , Diterpenes/therapeutic use , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Regulatory-Associated Protein of mTOR/metabolism , Animals , Arthritis, Gouty/metabolism , Cell Line , Diterpenes/chemistry , Gene Expression Regulation/drug effects , Inflammasomes/genetics , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , L-Lactate Dehydrogenase/antagonists & inhibitors , Leonurus/chemistry , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Structure , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Nigericin/toxicity , Phosphorylation , Regulatory-Associated Protein of mTOR/genetics , Up-Regulation
18.
J Biol Chem ; 297(1): 100775, 2021 07.
Article in English | MEDLINE | ID: mdl-34022218

ABSTRACT

Cellular pyruvate is an essential metabolite at the crossroads of glycolysis and oxidative phosphorylation, capable of supporting fermentative glycolysis by reduction to lactate mediated by lactate dehydrogenase (LDH) among other functions. Several inherited diseases of mitochondrial metabolism impact extracellular (plasma) pyruvate concentrations, and [1-13C]pyruvate infusion is used in isotope-labeled metabolic tracing studies, including hyperpolarized magnetic resonance spectroscopic imaging. However, how these extracellular pyruvate sources impact intracellular metabolism is not clear. Herein, we examined the effects of excess exogenous pyruvate on intracellular LDH activity, extracellular acidification rates (ECARs) as a measure of lactate production, and hyperpolarized [1-13C]pyruvate-to-[1-13C]lactate conversion rates across a panel of tumor and normal cells. Combined LDH activity and LDHB/LDHA expression analysis intimated various heterotetrameric isoforms comprising LDHA and LDHB in tumor cells, not only canonical LDHA. Millimolar concentrations of exogenous pyruvate induced substrate inhibition of LDH activity in both enzymatic assays ex vivo and in live cells, abrogated glycolytic ECAR, and inhibited hyperpolarized [1-13C]pyruvate-to-[1-13C]lactate conversion rates in cellulo. Of importance, the extent of exogenous pyruvate-induced inhibition of LDH and glycolytic ECAR in live cells was highly dependent on pyruvate influx, functionally mediated by monocarboxylate transporter-1 localized to the plasma membrane. These data provided evidence that highly concentrated bolus injections of pyruvate in vivo may transiently inhibit LDH activity in a tissue type- and monocarboxylate transporter-1-dependent manner. Maintaining plasma pyruvate at submillimolar concentrations could potentially minimize transient metabolic perturbations, improve pyruvate therapy, and enhance quantification of metabolic studies, including hyperpolarized [1-13C]pyruvate magnetic resonance spectroscopic imaging and stable isotope tracer experiments.


Subject(s)
L-Lactate Dehydrogenase/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Pyruvic Acid/pharmacology , Symporters/metabolism , Acids/metabolism , Buffers , Carbon Isotopes , Cell Extracts , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Extracellular Space/chemistry , Glycolysis/drug effects , Humans , Inhibitory Concentration 50 , Kinetics , L-Lactate Dehydrogenase/metabolism , Lactic Acid/biosynthesis , Substrate Specificity/drug effects
19.
J Med Chem ; 64(11): 7630-7645, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34009979

ABSTRACT

A class of quinazolone thiazoles was identified as new structural scaffolds for potential antibacterial conquerors to tackle dreadful resistance. Some prepared compounds exhibited favorable bacteriostatic efficiencies on tested bacteria, and the most representative 5j featuring the 4-trifluoromethylphenyl group possessed superior performances against Escherichia coli and Pseudomonas aeruginosa to norfloxacin. Further studies revealed that 5j with inappreciable hemolysis could hinder the formation of bacterial biofilms and trigger reactive oxygen species generation, which could take responsibility for emerging low resistance. Subsequent paralleled exploration discovered that 5j not only disintegrated outer and inner membranes to induce leakage of cytoplasmic contents but also broke the metabolism by suppressing dehydrogenase. Meanwhile, derivative 5j could intercalate into DNA to exert powerful antibacterial properties. Moreover, compound 5j gave synergistic effects against some Gram-negative bacteria in combination with norfloxacin. These findings indicated that this novel structural type of quinazolone thiazoles showed therapeutic foreground in struggling with Gram-negative bacterial infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Azoles/chemistry , Gram-Negative Bacteria/drug effects , Thiazoles/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Bacterial Outer Membrane/drug effects , Bacterial Outer Membrane/metabolism , Binding Sites , Binding, Competitive , Biofilms/drug effects , Crystallography, X-Ray , Drug Design , Drug Resistance, Bacterial/drug effects , Escherichia coli/physiology , Gram-Positive Bacteria/drug effects , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/metabolism , Microbial Sensitivity Tests , Molecular Conformation , Molecular Docking Simulation , Pseudomonas aeruginosa/physiology , Reactive Oxygen Species/metabolism , Thiazoles/metabolism , Thiazoles/pharmacology
20.
Br J Cancer ; 124(10): 1670-1679, 2021 05.
Article in English | MEDLINE | ID: mdl-33742144

ABSTRACT

BACKGROUND: Lactate dehydrogenase (LDH) is a critical metabolic enzyme. LDH A (LDHA) overexpression is a hallmark of aggressive malignancies and has been linked to tumour initiation, reprogramming and progression in multiple tumour types. However, successful LDHA inhibition strategies have not materialised in the translational and clinical space. We sought to develop a rational strategy for LDHA suppression in the context of solid tumour treatment. METHODS: We utilised a doxycycline-inducible short hairpin RNA (shRNA) system to generate LDHA suppression. Lactate and LDH activity levels were measured biochemically and kinetically using hyperpolarised 13C-pyruvate nuclear magnetic resonance spectroscopy. We evaluated effects of LDHA suppression on cellular proliferation and clonogenic survival, as well as on tumour growth, in orthotopic models of anaplastic thyroid carcinoma (ATC) and head and neck squamous cell carcinoma (HNSCC), alone or in combination with radiation. RESULTS: shRNA suppression of LDHA generated a time-dependent decrease in LDH activity with transient shifts in intracellular lactate levels, a decrease in carbon flux from pyruvate into lactate and compensatory shifts in metabolic flux in glycolysis and the Krebs cycle. LDHA suppression decreased cellular proliferation and temporarily stunted tumour growth in ATC and HNSCC xenografts but did not by itself result in tumour cure, owing to the maintenance of residual viable cells. Only when chronic LDHA suppression was combined with radiation was a functional cure achieved. CONCLUSIONS: Successful targeting of LDHA requires exquisite dose and temporal control without significant concomitant off-target toxicity. Combinatorial strategies with conventional radiation are feasible as long as the suppression is targeted, prolonged and non-toxic.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Head and Neck Neoplasms/drug therapy , L-Lactate Dehydrogenase/genetics , Molecular Targeted Therapy/methods , Squamous Cell Carcinoma of Head and Neck/drug therapy , Algorithms , Animals , Cell Line, Tumor , Down-Regulation/drug effects , Down-Regulation/genetics , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Feasibility Studies , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , L-Lactate Dehydrogenase/antagonists & inhibitors , Metabolomics , Mice , Mice, Nude , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...