Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 238
Filter
1.
Biochem Biophys Res Commun ; 584: 7-14, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34753066

ABSTRACT

Patients with triple-negative breast cancer have a poor prognosis as only a few efficient targeted therapies are available. Cancer cells are characterized by their unregulated proliferation and require large amounts of nucleotides to replicate their DNA. One-carbon metabolism contributes to purine and pyrimidine nucleotide synthesis by supplying one carbon atom. Although mitochondrial one-carbon metabolism has recently been focused on as an important target for cancer treatment, few specific inhibitors have been reported. In this study, we aimed to examine the effects of DS18561882 (DS18), a novel, orally active, specific inhibitor of methylenetetrahydrofolate dehydrogenase (MTHFD2), a mitochondrial enzyme involved in one-carbon metabolism. Treatment with DS18 led to a marked reduction in cancer-cell proliferation; however, it did not induce cell death. Combinatorial treatment with DS18 and inhibitors of checkpoint kinase 1 (Chk1), an activator of the S phase checkpoint pathway, efficiently induced apoptotic cell death in breast cancer cells and suppressed tumorigenesis in a triple-negative breast cancer patient-derived xenograft model. Mechanistically, MTHFD2 inhibition led to cell cycle arrest and slowed nucleotide synthesis. This finding suggests that DNA replication stress occurs due to nucleotide shortage and that the S-phase checkpoint pathway is activated, leading to cell-cycle arrest. Combinatorial treatment with both inhibitors released cell-cycle arrest, but induced accumulation of DNA double-strand breaks, leading to apoptotic cell death. Collectively, a combination of MTHFD2 and Chk1 inhibitors would be a rational treatment option for patients with triple-negative breast cancer.


Subject(s)
Aminohydrolases/antagonists & inhibitors , Checkpoint Kinase 1/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Methylenetetrahydrofolate Dehydrogenase (NADP)/antagonists & inhibitors , Multifunctional Enzymes/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Administration, Oral , Aminohydrolases/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Checkpoint Kinase 1/metabolism , Drug Therapy, Combination , Enzyme Inhibitors/administration & dosage , Female , Humans , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Multifunctional Enzymes/metabolism , S Phase Cell Cycle Checkpoints/drug effects , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays/methods
2.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Article in English | MEDLINE | ID: mdl-34544865

ABSTRACT

Bats are responsible for the zoonotic transmission of several major viral diseases, including those leading to the 2003 SARS outbreak and likely the ongoing COVID-19 pandemic. While comparative genomics studies have revealed characteristic adaptations of the bat innate immune system, functional genomic studies are urgently needed to provide a foundation for the molecular dissection of the viral tolerance in bats. Here we report the establishment of genome-wide RNA interference (RNAi) and CRISPR libraries for the screening of the model megabat, Pteropus alecto. We used the complementary RNAi and CRISPR libraries to interrogate P. alecto cells for infection with two different viruses: mumps virus and influenza A virus, respectively. Independent screening results converged on the endocytosis pathway and the protein secretory pathway as required for both viral infections. Additionally, we revealed a general dependence of the C1-tetrahydrofolate synthase gene, MTHFD1, for viral replication in bat cells and human cells. The MTHFD1 inhibitor, carolacton, potently blocked replication of several RNA viruses, including SARS-CoV-2. We also discovered that bats have lower expression levels of MTHFD1 than humans. Our studies provide a resource for systematic inquiry into the genetic underpinnings of bat biology and a potential target for developing broad-spectrum antiviral therapy.


Subject(s)
Aminohydrolases/genetics , COVID-19/genetics , Formate-Tetrahydrofolate Ligase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multienzyme Complexes/genetics , Pandemics , Aminohydrolases/antagonists & inhibitors , Animals , Antiviral Agents/therapeutic use , COVID-19/virology , Cell Line , Chiroptera/genetics , Chiroptera/virology , Formate-Tetrahydrofolate Ligase/antagonists & inhibitors , Humans , Methylenetetrahydrofolate Dehydrogenase (NADP)/antagonists & inhibitors , Minor Histocompatibility Antigens , Multienzyme Complexes/antagonists & inhibitors , RNA Viruses/genetics , SARS-CoV-2/pathogenicity , Virus Replication/genetics , COVID-19 Drug Treatment
3.
J Med Chem ; 64(15): 11288-11301, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34337952

ABSTRACT

Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) plays an important role in one-carbon metabolism. The MTHFD2 gene is upregulated in various cancers but very low or undetectable in normal proliferating cells, and therefore a potential target for cancer treatment. In this study, we present the structure of MTHFD2 in complex with xanthine derivative 15, which allosterically binds to MTHFD2 and coexists with the substrate analogue. A kinetic study demonstrated the uncompetitive inhibition of MTHFD2 by 15. Allosteric inhibitors often provide good selectivity and, indeed, xanthine derivatives are highly selective for MTHFD2. Moreover, several conformational changes were observed upon the binding of 15, which impeded the binding of the cofactor and phosphate to MTHFD2. To the best of our knowledge, this is the first study to identify allosteric inhibitors targeting the MTHFD family and our results would provide insights on the inhibition mechanism of MTHFD proteins and the development of novel inhibitors.


Subject(s)
Aminohydrolases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Methylenetetrahydrofolate Dehydrogenase (NADP)/antagonists & inhibitors , Multifunctional Enzymes/antagonists & inhibitors , Xanthine/pharmacology , Allosteric Site/drug effects , Aminohydrolases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Models, Molecular , Molecular Structure , Multifunctional Enzymes/metabolism , Structure-Activity Relationship , Xanthine/chemical synthesis , Xanthine/chemistry
4.
Nat Commun ; 12(1): 1940, 2021 03 29.
Article in English | MEDLINE | ID: mdl-33782411

ABSTRACT

Metabolic enzymes and metabolites display non-metabolic functions in immune cell signalling that modulate immune attack ability. However, whether and how a tumour's metabolic remodelling contributes to its immune resistance remain to be clarified. Here we perform a functional screen of metabolic genes that rescue tumour cells from effector T cell cytotoxicity, and identify the embryo- and tumour-specific folate cycle enzyme methylenetetrahydrofolate dehydrogenase 2 (MTHFD2). Mechanistically, MTHFD2 promotes basal and IFN-γ-stimulated PD-L1 expression, which is necessary for tumourigenesis in vivo. Moreover, IFN-γ stimulates MTHFD2 through the AKT-mTORC1 pathway. Meanwhile, MTHFD2 drives the folate cycle to sustain sufficient uridine-related metabolites including UDP-GlcNAc, which promotes the global O-GlcNAcylation of proteins including cMYC, resulting in increased cMYC stability and PD-L1 transcription. Consistently, the O-GlcNAcylation level positively correlates with MTHFD2 and PD-L1 in pancreatic cancer patients. These findings uncover a non-metabolic role for MTHFD2 in cell signalling and cancer biology.


Subject(s)
Aminohydrolases/genetics , B7-H1 Antigen/genetics , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multifunctional Enzymes/genetics , Pancreatic Neoplasms/genetics , Protein Processing, Post-Translational , T-Lymphocytes, Cytotoxic/immunology , Aminohydrolases/antagonists & inhibitors , Aminohydrolases/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Carcinogenesis/immunology , Carcinogenesis/pathology , Cell Line, Tumor , Embryo, Mammalian , Fibroblasts/immunology , Fibroblasts/pathology , Folic Acid/immunology , Folic Acid/metabolism , Humans , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/immunology , Methylenetetrahydrofolate Dehydrogenase (NADP)/antagonists & inhibitors , Methylenetetrahydrofolate Dehydrogenase (NADP)/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Multifunctional Enzymes/antagonists & inhibitors , Multifunctional Enzymes/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Primary Cell Culture , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Signal Transduction , T-Lymphocytes, Cytotoxic/pathology , Tumor Burden , Tumor Escape , Uridine Diphosphate N-Acetylglucosamine/metabolism , Xenograft Model Antitumor Assays
5.
Theranostics ; 11(8): 3661-3675, 2021.
Article in English | MEDLINE | ID: mdl-33664854

ABSTRACT

Rationale: Lung adenocarcinoma (LUAD) is an aggressive disease with high propensity of metastasis. Among patients with early-stage disease, more than 30% of them may relapse or develop metastasis. There is an unmet medical need to stratify patients with early-stage LUAD according to their risk of relapse/metastasis to guide preventive or therapeutic approaches. In this study, we identified 4 genes that can serve both therapeutic and diagnostic (theranostic) purposes. Methods: Three independent datasets (GEO, TCGA, and KMPlotter) were used to evaluate gene expression profile of patients with LUAD by unbiased screening approach. Upon significant genes uncovered, functional enrichment analysis was carried out. The predictive power of their expression on patient prognosis were evaluated. Once confirmed their theranostic roles by integrated bioinformatics, we further conducted in vitro and in vivo validation. Results: We found that four genes (ADAM9, MTHFD2, RRM2, and SLC2A1) were associated with poor patient outcomes with an increased hazard ratio in LUAD. Knockdown of them, both separately and simultaneously, suppressed lung cancer cell proliferation and migration ability in vitro and prolonged survival time in metastatic tumor mouse models. Moreover, these four biomarkers were found to be overexpressed in tumor tissues from LUAD patients, and the total immunohistochemical staining scores correlated with poor prognosis. Conclusions: These results suggest that these four identified genes could be theranostic biomarkers for stratifying high-risk patients who develop relapse/metastasis in early-stage LUAD. Developing therapeutic approaches for the four biomarkers may benefit early-stage LUAD patients after surgery.


Subject(s)
Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/secondary , Biomarkers, Tumor/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , A549 Cells , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/genetics , Adenocarcinoma of Lung/surgery , Aminohydrolases/antagonists & inhibitors , Aminohydrolases/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Knockdown Techniques , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/genetics , HEK293 Cells , Humans , Lung Neoplasms/surgery , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/antagonists & inhibitors , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Mice , Mice, SCID , Multifunctional Enzymes/antagonists & inhibitors , Multifunctional Enzymes/genetics , Precision Medicine , Prognosis , Ribonucleoside Diphosphate Reductase/antagonists & inhibitors , Ribonucleoside Diphosphate Reductase/genetics , Risk Factors , Transcriptome , Xenograft Model Antitumor Assays
6.
Drug Discov Today ; 26(3): 817-825, 2021 03.
Article in English | MEDLINE | ID: mdl-33316375

ABSTRACT

Folate-mediated one-carbon metabolism (FOCM) supports vital events for the growth and survival of proliferating cells. Nucleotide synthesis and DNA methylation are the biochemical bases of cancers that are highly dependent on FOCM. Recent studies revealed that FOCM is connected with redox homeostasis and epigenetics in cancer. Furthermore, folate-metabolizing enzymes, such as serine hydroxymethyltransferase 2 (SHMT2) and methylenetetrahydrofolate dehydrogenase 2 (MTHFD2), are associated with the development of cancers, including breast cancer, highlighting their potential application in tumor-targeted therapy. Therefore, targeting metabolizing enzymes, especially SHMT2 and MTHFD2, provides a novel strategy for cancer treatment. In this review, we outline current understanding of the functions of SHMT2 and MTHFD2, discussing their expression, potential functions, and regulatory mechanism in cancers. Furthermore, we discuss examples of inhibitors of SHMT2 and MTHFD2.


Subject(s)
Molecular Targeted Therapy , Neoplasms/drug therapy , Aminohydrolases/antagonists & inhibitors , Aminohydrolases/metabolism , Animals , Carbon/metabolism , Cell Survival/physiology , DNA Methylation/genetics , Folic Acid/metabolism , Glycine Hydroxymethyltransferase/antagonists & inhibitors , Glycine Hydroxymethyltransferase/metabolism , Humans , Methylenetetrahydrofolate Dehydrogenase (NADP)/antagonists & inhibitors , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Multifunctional Enzymes/antagonists & inhibitors , Multifunctional Enzymes/metabolism , Neoplasms/pathology
7.
Mol Cancer Ther ; 19(11): 2245-2255, 2020 11.
Article in English | MEDLINE | ID: mdl-32879053

ABSTRACT

One-carbon (1C) metabolism encompasses folate-mediated 1C transfer reactions and related processes, including nucleotide and amino acid biosynthesis, antioxidant regeneration, and epigenetic regulation. 1C pathways are compartmentalized in the cytosol, mitochondria, and nucleus. 1C metabolism in the cytosol has been an important therapeutic target for cancer since the inception of modern chemotherapy, and "antifolates" targeting cytosolic 1C pathways continue to be a mainstay of the chemotherapy armamentarium for cancer. Recent insights into the complexities of 1C metabolism in cancer cells, including the critical role of the mitochondrial 1C pathway as a source of 1C units, glycine, reducing equivalents, and ATP, have spurred the discovery of novel compounds that target these reactions, with particular focus on 5,10-methylene tetrahydrofolate dehydrogenase 2 and serine hydroxymethyltransferase 2. In this review, we discuss key aspects of 1C metabolism, with emphasis on the importance of mitochondrial 1C metabolism to metabolic homeostasis, its relationship with the oncogenic phenotype, and its therapeutic potential for cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Energy Metabolism/drug effects , Metabolic Networks and Pathways/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Targeted Therapy , Neoplasms/metabolism , Aminohydrolases/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Biomarkers , Carbon/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glycine Hydroxymethyltransferase/antagonists & inhibitors , Humans , Methylenetetrahydrofolate Dehydrogenase (NADP)/antagonists & inhibitors , Molecular Targeted Therapy/methods , Multifunctional Enzymes/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/pathology , Serine/biosynthesis
8.
J Med Chem ; 62(22): 10204-10220, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31638799

ABSTRACT

We report the discovery of a potent and isozyme-selective MTHFD2 inhibitor, DS18561882 (2). Through investigation of the substituents on our tricyclic coumarin scaffold (1,2,3,4-tetrahydrochromeno[3,4-c]pyridin-5-one), MTHFD2 inhibitory activity was shown to be elevated by incorporating an amine moiety at the 8-position and a methyl group at the 7-position of the initial lead 1. X-ray structure analysis revealed that a key interaction for enhanced potency was salt bridge formation between the amine moiety and the diphosphate linker of an NAD+ cofactor. Furthermore, ortho-substituted sulfonamide in place of benzoic acid of 1 significantly improved cell permeability and cell-based growth inhibition against a human breast cancer cell line. The thus-optimized DS18561882 showed the strongest cell-based activity (GI50 = 140 nM) in the class, a good oral pharmacokinetic profile, and thereby tumor growth inhibition in a mouse xenograft model upon oral administration.


Subject(s)
Aminohydrolases/antagonists & inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Methylenetetrahydrofolate Dehydrogenase (NADP)/antagonists & inhibitors , Multifunctional Enzymes/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Crystallography, X-Ray , Female , Humans , Male , Mice, Inbred BALB C , Structure-Activity Relationship , Xenograft Model Antitumor Assays
9.
J Natl Cancer Inst ; 111(6): 584-596, 2019 Jun 01.
Article in English | MEDLINE | ID: mdl-30534944

ABSTRACT

BACKGROUND: Overcoming oxidative stress is a critical step for tumor progression; however, the underlying mechanisms in colorectal cancer (CRC) remain unclear. METHODS: We investigated nicotinamide adenine dinucleotide (phosphate) (NAD(P))-dependent enzyme methylene tetrahydrofolate dehydrogenase 2 (MTHFD2) expression, clinical relevance, redox modification, and molecular mechanisms using the CRC cells and tissues (n = 462 paired samples). The antitumor effects of MTHFD2 inhibitor LY345899 on CRC tumorigenesis and metastasis were evaluated in vitro and in vivo. Data analysis used Kaplan-Meier, Pearson's correlation, and Student t test where appropriate. All statistical tests were two-sided. RESULTS: Here, we report that the patients with high expression of MTHFD2 have a shorter overall survival (HR = 1.62, 95% CI = 1.12 to 2.36, P = .01) and disease-free survival (HR = 1.55, 95% CI = 1.07 to 2.27, P = .02) than patients with low MTHFD2 expression. Suppression of MTHFD2 disturbs NADPH and redox homeostasis and accelerates cell death under oxidative stress, such as hypoxia or anchorage independence (P ≤ .01 for all). Also, genetic or pharmacological inhibition of MTHFD2 suppresses CRC cell growth and lung and peritoneal metastasis in cell-based xenografts (n = 5-8 mice per group). Importantly, LY345899 treatment statistically significantly suppresses tumor growth and decreases the tumor weight in CRC patient-derived xenograft models (n = 10 mice per group, mean [SD] tumor weight of the vehicle-treated group was 1.83 [0.19] mg vs 0.74 [0.30] mg for the LY345899-treated group, P < .001). CONCLUSIONS: Our study presents evidence that MTHFD2 confers redox homeostasis and promotes CRC cell growth and metastasis. The folate analog LY345899 as MTHFD2 inhibitor displays therapeutic activity against CRC and warrants further clinical investigation for CRC treatment.


Subject(s)
Aminohydrolases/antagonists & inhibitors , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Enzyme Inhibitors/pharmacology , Glutamates/pharmacology , Methylenetetrahydrofolate Dehydrogenase (NADP)/antagonists & inhibitors , Multifunctional Enzymes/antagonists & inhibitors , Aminohydrolases/genetics , Aminohydrolases/metabolism , Animals , Anoikis/drug effects , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Female , Humans , Lung Neoplasms/secondary , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Random Allocation , Signal Transduction , Transcription, Genetic , Xenograft Model Antitumor Assays
10.
Cell Physiol Biochem ; 51(2): 991-1000, 2018.
Article in English | MEDLINE | ID: mdl-30466107

ABSTRACT

BACKGROUND/AIMS: To investigate the role of methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) in the clinical prognosis and cell biology of renal cell carcinoma (RCC). METHODS: A total of 137 RCC tissues were evaluated by immunohistochemistry. The relationship between MTHFD2 overexpression and clinical parameters and vimentin expression was assessed. Kaplan-Meier curves and the log-rank test were applied for survival analysis according to MTHFD2 and vimentin expression in RCC tissues. The expression of MTHFD2 mRNA and protein was examined by quantitative reverse transcription PCR and western blotting, respectively. To determine further the biological activity of MTHFD2 in RCC, 786-O cells were transfected with short hairpin RNA specifically targeting MTHFD2 (shMTHFD2) with or without tumor necrosis factor (TNF)-α stimulation. Cell proliferation, cell migration and invasion and drug sensitivity were subsequently assessed using Cell Counting Kit-8, wound healing, and Transwell assays. RESULTS: Immunohistochemical analysis demonstrated that both MTHFD2 and vimentin overexpression was positively associated with clinical staging, pathological grade, and poor overall survival (all P < 0.05). MTHFD2 expression was closely correlated with vimentin overexpression in RCC (r = 0.402, P < 0.001). After knocking down MTHFD2 expression in 786-O cells, decreased cell proliferation, migration, and invasion were observed and accompanied by the reduced expression of vimentin. The effects of MTHFD2 down-regulation could be partially restrained by TNF-α treatment. Vimentin expression and cell migration and invasion, but not cell proliferation, were reversed by TNF-α stimulation. Furthermore, treatment of 786-O cells with shMTHFD2 increased their sensitivity to chemotherapy drugs. CONCLUSION: The current results demonstrated that MTHFD2 was overexpressed in RCC and associated with poor clinical characteristics, vimentin expression, and cellular features connected to malignant disease, thus, implicating MTHFD2 as a potential target for RCC therapy.


Subject(s)
Aminohydrolases/metabolism , Carcinoma, Renal Cell/pathology , Cell Proliferation , Kidney Neoplasms/pathology , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Multifunctional Enzymes/metabolism , Vimentin/metabolism , Aged , Aminohydrolases/antagonists & inhibitors , Aminohydrolases/genetics , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/mortality , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Female , Humans , Kaplan-Meier Estimate , Kidney Neoplasms/mortality , Male , Methylenetetrahydrofolate Dehydrogenase (NADP)/antagonists & inhibitors , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Middle Aged , Multifunctional Enzymes/antagonists & inhibitors , Multifunctional Enzymes/genetics , Neoplasm Grading , Prognosis , RNA Interference , RNA, Small Interfering/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Vimentin/genetics
11.
Proc Natl Acad Sci U S A ; 114(12): E2319-E2326, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28265077

ABSTRACT

Arsenic exposure increases risk for cancers and is teratogenic in animal models. Here we demonstrate that small ubiquitin-like modifier (SUMO)- and folate-dependent nuclear de novo thymidylate (dTMP) biosynthesis is a sensitive target of arsenic trioxide (As2O3), leading to uracil misincorporation into DNA and genome instability. Methylenetetrahydrofolate dehydrogenase 1 (MTHFD1) and serine hydroxymethyltransferase (SHMT) generate 5,10-methylenetetrahydrofolate for de novo dTMP biosynthesis and translocate to the nucleus during S-phase, where they form a multienzyme complex with thymidylate synthase (TYMS) and dihydrofolate reductase (DHFR), as well as the components of the DNA replication machinery. As2O3 exposure increased MTHFD1 SUMOylation in cultured cells and in in vitro SUMOylation reactions, and increased MTHFD1 ubiquitination and MTHFD1 and SHMT1 degradation. As2O3 inhibited de novo dTMP biosynthesis in a dose-dependent manner, increased uracil levels in nuclear DNA, and increased genome instability. These results demonstrate that MTHFD1 and SHMT1, which are key enzymes providing one-carbon units for dTMP biosynthesis in the form of 5,10-methylenetetrahydrofolate, are direct targets of As2O3-induced proteolytic degradation, providing a mechanism for arsenic in the etiology of cancer and developmental anomalies.


Subject(s)
Aminohydrolases/antagonists & inhibitors , Cell Nucleus/metabolism , Formate-Tetrahydrofolate Ligase/antagonists & inhibitors , Methylenetetrahydrofolate Dehydrogenase (NADP)/antagonists & inhibitors , Multienzyme Complexes/antagonists & inhibitors , Oxides/toxicity , Small Ubiquitin-Related Modifier Proteins/antagonists & inhibitors , Thymidine Monophosphate/biosynthesis , Aminohydrolases/genetics , Aminohydrolases/metabolism , Animals , Arsenic Trioxide , Arsenicals , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Cell Nucleus/genetics , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/metabolism , Formate-Tetrahydrofolate Ligase/genetics , Formate-Tetrahydrofolate Ligase/metabolism , Genomic Instability/drug effects , Glycine Hydroxymethyltransferase/genetics , Glycine Hydroxymethyltransferase/metabolism , Humans , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mice , Mice, Knockout , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Proteolysis , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Uracil/metabolism
12.
J Biomol Struct Dyn ; 35(2): 287-299, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26725317

ABSTRACT

Brucella melitensis is a pathogenic Gram-negative bacterium which is known for causing zoonotic diseases (Brucellosis). The organism is highly contagious and has been reported to be used as bioterrorism agent against humans. Several antibiotics and vaccines have been developed but these antibiotics have exhibited the sign of antibiotic resistance or ineffective at lower concentrations, which imposes an urgent need to identify the novel drugs/drug targets against this organism. In this work, metabolic pathways analysis has been performed with different filters such as non-homology with humans, essentially of genes and choke point analysis, leading to identification of novel drug targets. A total of 18 potential drug target proteins were filtered out and used to develop the high confidence protein-protein interaction network The Phosphoribosyl-AMP cyclohydrolase (HisI) protein has been identified as potential drug target on the basis of topological parameters. Further, a homology model of (HisI) protein has been developed using Modeller with multiple template (1W6Q (48%), 1ZPS (55%), and 2ZKN (48%)) approach and validated using PROCHECK and Verify3D. The virtual high throughput screening (vHTS) using DockBlaster tool has been performed against 16,11,889 clean fragments from ZINC database. Top 500 molecules from DockBlaster were docked using Vina. The docking analysis resulted in ZINC04880153 showing the lowest binding energy (-9.1 kcal/mol) with the drug target. The molecular dynamics study of the complex HisI-ZINC04880153 was conducted to analyze the stability and fluctuation of ligand within the binding pocket of HisI. The identified ligand could be analyzed in the wet-lab based experiments for future drug discovery.


Subject(s)
Aminohydrolases/chemistry , Aminohydrolases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Brucella melitensis/metabolism , Enzyme Inhibitors/chemistry , Metabolic Networks and Pathways , Aminohydrolases/antagonists & inhibitors , Bacterial Proteins/pharmacology , Binding Sites , Computational Biology/methods , Drug Discovery , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Bonding , Models, Molecular , Molecular Conformation , Molecular Dynamics Simulation , Protein Binding , Protein Interaction Mapping , Protein Interaction Maps
13.
Appl Microbiol Biotechnol ; 100(11): 4779-89, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27094187

ABSTRACT

Pterin deaminase is an amidohydrolase enzyme hydrolyzing pteridines to form lumazine derivatives and ammonia. The enzyme captured the attention of scientists as early as 1959 and had been patented for its application as an anticancer agent. It is ubiquitously present in prokaryotes and has been reported in some eukaryotes such as honey bee, silkworm and rats. The enzyme has been observed to have a spectrum of substrates with the formation of respective lumazines. The role of the substrates of the enzyme in various metabolic pathways warrants a significant role in the biological activity of both prokaryotes and eukaryotes. Even though the functions of the enzyme have been explored in prokaryotes, their niche in the eukaryotic system is not clear. There is very few information on the structural and functional properties of the enzyme. This review has been congregated to emphasize the significance of pterin deaminase and analyzes the lacunae in understanding the biological characters of the enzyme.


Subject(s)
Amidohydrolases/metabolism , Aminohydrolases/metabolism , Aminohydrolases/antagonists & inhibitors , Animals , Biopterins/analogs & derivatives , Biopterins/metabolism , Eukaryotic Cells/enzymology , Prokaryotic Cells/enzymology , Pteridines/chemistry
14.
BMC Biochem ; 17: 4, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26956223

ABSTRACT

BACKGROUND: Pseudomonas aeruginosa, a Gram-negative, aerobic coccobacillus bacterium is an opportunistic human pathogen and worldwide the fourth most common cause of hospital-acquired infections which are often high mortality such as ventilator-associated pneumoniae. The polyamine metabolism of P. aeruginosa and particularly the deacetylation of acetylpolyamines has been little studied up to now. Results with other bacterial pathogens e.g., Y. pestis suggest that polyamines may be involved in the formation of biofilms or confer resistance against certain antibiotics. RESULTS: To elucidate the role of acetylpolyamines and their enzymatic deacetylation in more detail, all three putative acetylpolyamine amidohydrolases (APAHs) from P. aeruginosa have been expressed in enzymatic active form. The APAHs PA0321 and PA1409 are shown to be true polyamine deacetylases, whereas PA3774 is not able to deacetylate acetylated polyamines. Every APAH can hydrolyze trifluoroacetylated lysine-derivatives, but only PA1409 and much more efficiently PA3774 can also process the plain acetylated lysine substrate. P. aeruginosa is able to utilize acetylcadaverine and acetylputrescine as a carbon source under glucose starvation. If either the PA0321 or the PA1409 but not the PA3774 gene is disrupted, the growth of P. aeruginosa is reduced and delayed. In addition, we were able to show that the APAH inhibitors SAHA and SATFMK induce biofilm formation in both PA14 and PAO1 wildtype strains. CONCLUSIONS: P. aeruginosa has two functional APAHs, PA0321 and PA1409 which enable the utilization of acetylpolyamines for the metabolism of P. aeruginosa. In contrast, the physiological role of the predicted APAH, PA3774, remains to be elucidated. Its ability to deacetylate synthetic acetylated lysine substrates points to a protein deacetylation functionality with yet unknown substrates.


Subject(s)
Aminohydrolases/metabolism , Pseudomonas aeruginosa/enzymology , Amino Acid Sequence , Aminohydrolases/antagonists & inhibitors , Biofilms/drug effects , Histone Deacetylase 6 , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/chemistry , Humans , Molecular Sequence Data , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Sequence Alignment , Substrate Specificity
15.
Biochemistry ; 54(30): 4692-703, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26200446

ABSTRACT

Polyamines are essential aliphatic polycations that bind to nucleic acids and accordingly are involved in a variety of cellular processes. Polyamine function can be regulated by acetylation and deacetylation, just as histone function can be regulated by lysine acetylation and deacetylation. Acetylpolyamine amidohydrolase (APAH) from Mycoplana ramosa is a zinc-dependent polyamine deacetylase that shares approximately 20% amino acid sequence identity with human histone deacetylases. We now report the X-ray crystal structures of APAH-inhibitor complexes in a new and superior crystal form that diffracts to very high resolution (1.1-1.4 Å). Inhibitors include previously synthesized analogues of N(8)-acetylspermidine bearing trifluoromethylketone, thiol, and hydroxamate zinc-binding groups [Decroos, C., Bowman, C. M., and Christianson, D. W. (2013) Bioorg. Med. Chem. 21, 4530], and newly synthesized hydroxamate analogues of shorter, monoacetylated diamines, the most potent of which is the hydroxamate analogue of N-acetylcadaverine (IC50 = 68 nM). The high-resolution crystal structures of APAH-inhibitor complexes provide key inferences about the inhibition and catalytic mechanism of zinc-dependent deacetylases. For example, the trifluoromethylketone analogue of N(8)-acetylspermidine binds as a tetrahedral gem-diol that mimics the tetrahedral intermediate and its flanking transition states in catalysis. Surprisingly, this compound is also a potent inhibitor of human histone deacetylase 8 with an IC50 of 260 nM. Crystal structures of APAH-inhibitor complexes are determined at the highest resolution of any currently existing zinc deacetylase structure and thus represent the most accurate reference points for understanding structure-mechanism and structure-inhibition relationships in this critically important enzyme family.


Subject(s)
Aminohydrolases/chemistry , Bacterial Proteins/chemistry , Brucellaceae/enzymology , Enzyme Inhibitors/chemistry , Zinc/chemistry , Aminohydrolases/antagonists & inhibitors , Bacterial Proteins/antagonists & inhibitors , Crystallography, X-Ray , Humans , Protein Structure, Tertiary
16.
Anal Bioanal Chem ; 406(20): 4889-97, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24871864

ABSTRACT

High-throughput assays for drug screening applications have to fulfill particular specifications. Besides the capability to identify even compounds with low potency, one of the major issues is to minimize the number of false-positive hits in a screening campaign in order to reduce the logistic effort for the subsequent cherry picking and confirmation procedure. In this respect, fluorescence lifetime (FLT) appears as an ideal readout parameter that is supposed to be robust against autofluorescent and light-absorbing compounds, the most common source of systematic false positives. The extraordinary fluorescence features of the recently discovered [1,3]dioxolo[4,5-f][1,3] benzodioxole dyes were exploited to develop an FLT-based binding assay with exceptionally robust readout. The assay setup was comprehensively validated and shown to comply not only with all requirements for a powerful high-throughput screening assay but also to be suitable to determine accurate binding constants for inhibitors against enzymes of the histone deacetylase family. Using the described binding assay, the first inhibitors against three members of this enzyme family from Pseudomonas aeruginosa were identified. The compounds were characterized in terms of potency and selectivity profile. The novel ligand probe should also be applicable to other homologues of the histone deacetylase family that are inhibited by N-hydroxy-N'-phenyloctandiamide.


Subject(s)
Aminohydrolases/metabolism , Biological Assay , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Fluorescent Dyes/chemistry , High-Throughput Screening Assays , Pseudomonas aeruginosa/enzymology , Aminohydrolases/antagonists & inhibitors , Binding, Competitive , Fluorescence , Fluorescence Resonance Energy Transfer , Kinetics , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development
17.
Cell Host Microbe ; 14(4): 411-21, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-24139399

ABSTRACT

Several human APOBEC3 deaminases can inhibit HIV-1 replication in vitro. HIV-1 Vif counteracts this restriction by targeting APOBEC3 for proteasomal degradation. Human APOBEC3H (A3H) is highly polymorphic, with natural variants differing considerably in anti-HIV-1 activity in vitro. To examine HIV-1 adaptation to variation in A3H activity in a natural infection context, we determined the A3H haplotypes and Vif sequences from 76 recently infected HIV-1 patients. We detected A3H-specific Vif changes suggesting viral adaptation. The patient-derived Vif sequences were used to engineer viruses that specifically differed in their ability to counteract A3H. Replication of these Vif-variant viruses in primary T cells naturally expressing active or inactive A3H haplotypes showed that endogenously expressed A3H restricts HIV-1 replication. Proviral DNA from A3H-restricted viruses showed high levels of G-to-A mutations in an A3H-specific GA dinucleotide context. Taken together, our data validate A3H expressed at endogenous levels as a bona fide HIV-1 restriction factor.


Subject(s)
Adaptation, Biological , Aminohydrolases/antagonists & inhibitors , HIV-1/immunology , HIV-1/physiology , vif Gene Products, Human Immunodeficiency Virus/genetics , vif Gene Products, Human Immunodeficiency Virus/metabolism , Aminohydrolases/immunology , Cells, Cultured , DNA Mutational Analysis , DNA, Viral/chemistry , DNA, Viral/genetics , HIV Infections/virology , HIV-1/genetics , HIV-1/isolation & purification , Haplotypes , Host-Pathogen Interactions , Humans , Proviruses/genetics , Reverse Genetics , Selection, Genetic , Sequence Analysis, DNA , T-Lymphocytes/virology , Virus Replication
18.
Bioorg Med Chem ; 21(15): 4530-40, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23790721

ABSTRACT

Polyamines are small essential polycations involved in many biological processes. Enzymes of polyamine metabolism have been extensively studied and are attractive drug targets. Nevertheless, the reversible acetylation of polyamines remains poorly understood. Although eukaryotic N(8)-acetylspermidine deacetylase activity has already been detected and studied, the specific enzyme responsible for this activity has not yet been identified. However, a zinc deacetylase from Mycoplana ramosa, acetylpolyamine amidohydrolase (APAH), has been reported to use various acetylpolyamines as substrates. The recently solved crystal structure of this polyamine deacetylase revealed the formation of an 'L'-shaped active site tunnel at the dimer interface, with ideal dimensions and electrostatic properties for accommodating narrow, flexible, cationic polyamine substrates. Here, we report the design, synthesis, and evaluation of N(8)-acetylspermidine analogues bearing different zinc binding groups as potential inhibitors of APAH. Most of the synthesized compounds exhibit modest potency, with IC50 values in the mid-micromolar range, but compounds bearing hydroxamate or trifluoromethylketone zinc binding groups exhibit enhanced inhibitory potency in the mid-nanomolar range. These inhibitors will enable future explorations of acetylpolyamine function in both prokaryotes and eukaryotes.


Subject(s)
Aminohydrolases/antagonists & inhibitors , Spermidine/analogs & derivatives , Aminohydrolases/chemistry , Polyamines/metabolism , Spermidine/chemical synthesis , Spermidine/chemistry
19.
ChemMedChem ; 6(1): 81-8, 2011 Jan 03.
Article in English | MEDLINE | ID: mdl-20979083

ABSTRACT

C-Alkyl amidine analogues of asymmetric N(ω),N(ω)-dimethyl-L-arginine are dual-targeted inhibitors of both human DDAH-1 and nitric oxide (NO) synthase, and provide a promising scaffold for the development of therapeutics to control NO overproduction in a variety of pathologies including septic shock and some cancers. Using a two-part click-chemistry-mediated activity probe, a homologated series of C-alkyl amidines were ranked for their ability to inhibit DDAH-1 within cultured HEK 293T cells. N5-(1-Iminopentyl)-L-ornithine was determined to be the most potent compound in vitro (K(d)=7 µM) as well as in cultured cells, and the binding conformation and covalent reversible mode of inhibition was investigated by comparison of interactions made with DDAH-1 and a catalytically inactive C274S variant, as gauged by X-ray crystallography and isothermal titration calorimetry. By interrupting the ability of the inhibitor to form a covalent bond, the contribution of this interaction could be estimated. These results suggest that further stabilization of the covalent adduct is a promising strategy for lead optimization in the design of effective reagents to block NO synthesis.


Subject(s)
Amidines , Aminohydrolases/antagonists & inhibitors , Arginine/analogs & derivatives , Enzyme Inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Amidines/pharmacology , Aminohydrolases/genetics , Aminohydrolases/metabolism , Arginine/metabolism , Biological Availability , Calorimetry, Differential Scanning , Cell Line , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism
20.
Article in English | MEDLINE | ID: mdl-20047516

ABSTRACT

The nitrilase produced from a new isolate is evaluated for its activity in presence of a number of different ions and compounds at optimal conditions. It was found that the activity of nitrilase increased up to 10-20% in presence of most of the divalent ions at a concentration of 5 mM relative to the control. Silver, mercury, tin, DTT, ascorbic acid and thiourea, respectively, were observed as potential inhibitors of the enzyme catalysis. The investigation on storage stability of whole cells in presence of a number of stabilizers showed that the enzyme is stable (relative activity 50%) for more than 120 days at various temperatures.


Subject(s)
Aminohydrolases/metabolism , Cations, Divalent/metabolism , Metals/metabolism , Nitriles/metabolism , Streptomyces/enzymology , Aminohydrolases/antagonists & inhibitors , Aminohydrolases/chemistry , Biodegradation, Environmental , Cations, Divalent/chemistry , Cations, Divalent/pharmacology , Industrial Microbiology , Metals/chemistry , Metals/pharmacology , Protein Stability/drug effects , Sanitary Engineering/methods , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL