1
|
Fernández-Ramos D, Lopitz-Otsoa F, Lu SC, Mato JM. S-Adenosylmethionine: A Multifaceted Regulator in Cancer Pathogenesis and Therapy. Cancers (Basel) 2025; 17:535. [PMID: 39941901 PMCID: PMC11816870 DOI: 10.3390/cancers17030535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
S-adenosylmethionine (SAMe) is a key methyl donor that plays a critical role in a variety of cellular processes, such as DNA, RNA and protein methylation, essential for maintaining genomic stability, regulating gene expression and maintaining cellular homeostasis. The involvement of SAMe in cancer pathogenesis is multifaceted, as through its multiple cellular functions, it can influence tumor initiation, progression and therapeutic resistance. In addition, the connection of SAMe with polyamine synthesis and oxidative stress management further underscores its importance in cancer biology. Recent studies have highlighted the potential of SAMe as a biomarker for cancer diagnosis and prognosis. Furthermore, the therapeutic implications of SAMe are promising, with evidence suggesting that SAMe supplementation or modulation could improve the efficacy of existing cancer treatments by restoring proper methylation patterns and mitigating oxidative damage and protect against damage induced by chemotherapeutic drugs. Moreover, targeting methionine cycle enzymes to both regulate SAMe availability and SAMe-independent regulatory effects, particularly in methionine-dependent cancers such as colorectal and lung cancer, presents a promising therapeutic approach. Additionally, exploring epitranscriptomic regulations, such as m6A modifications, and their interaction with non-coding RNAs could enhance our understanding of tumor progression and resistance mechanisms. Precision medicine approaches integrating patient subtyping and combination therapies with chemotherapeutics, such as decitabine or doxorubicin, together with SAMe, can enhance chemosensitivity and modulate epigenomics, showing promising results that may improve treatment outcomes. This review comprehensively examines the various roles of SAMe in cancer pathogenesis, its potential as a diagnostic and prognostic marker, and its emerging therapeutic applications. While SAMe modulation holds significant promise, challenges such as bioavailability, patient stratification and context-dependent effects must be addressed before clinical implementation. In addition, better validation of the obtained results into specific cancer animal models would also help to bridge the gap between research and clinical practice.
Collapse
Affiliation(s)
- David Fernández-Ramos
- Precision Medicine and Metabolism Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (D.F.-R.); (F.L.-O.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Fernando Lopitz-Otsoa
- Precision Medicine and Metabolism Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (D.F.-R.); (F.L.-O.)
| | - Shelly C. Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - José M. Mato
- Precision Medicine and Metabolism Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (D.F.-R.); (F.L.-O.)
| |
Collapse
|
2
|
Bedard GT, Gilaj N, Peregrina K, Brew I, Tosti E, Shaffer K, Tyler PC, Edelmann W, Augenlicht LH, Schramm VL. Combined inhibition of MTAP and MAT2a mimics synthetic lethality in tumor models via PRMT5 inhibition. J Biol Chem 2024; 300:105492. [PMID: 38000655 PMCID: PMC10770533 DOI: 10.1016/j.jbc.2023.105492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Homozygous 5'-methylthioadenosine phosphorylase (MTAP) deletions occur in approximately 15% of human cancers. Co-deletion of MTAP and methionine adenosyltransferase 2 alpha (MAT2a) induces a synthetic lethal phenotype involving protein arginine methyltransferase 5 (PRMT5) inhibition. MAT2a inhibitors are now in clinical trials for genotypic MTAP-/- cancers, however the MTAP-/- genotype represents fewer than 2% of human colorectal cancers (CRCs), limiting the utility of MAT2a inhibitors in these and other MTAP+/+ cancers. Methylthio-DADMe-immucillin-A (MTDIA) is a picomolar transition state analog inhibitor of MTAP that renders cells enzymatically MTAP-deficient to induce the MTAP-/- phenotype. Here, we demonstrate that MTDIA and MAT2a inhibitor AG-270 combination therapy mimics synthetic lethality in MTAP+/+ CRC cell lines with similar effects in mouse xenografts and without adverse histology on normal tissues. Combination treatment is synergistic with a 104-fold increase in drug potency for inhibition of CRC cell growth in culture. Combined MTDIA and AG-270 decreases S-adenosyl-L-methionine and increases 5'-methylthioadenosine in cells. The increased intracellular methylthioadenosine:S-adenosyl-L-methionine ratio inhibits PRMT5 activity, leading to cellular arrest and apoptotic cell death by causing MDM4 alternative splicing and p53 activation. Combination MTDIA and AG-270 treatment differs from direct inhibition of PRMT5 by GSK3326595 by avoiding toxicity caused by cell death in the normal gut epithelium induced by the PRMT5 inhibitor. The combination of MTAP and MAT2a inhibitors expands this synthetic lethal approach to include MTAP+/+ cancers, especially the remaining 98% of CRCs without the MTAP-/- genotype.
Collapse
Affiliation(s)
- Gabriel T Bedard
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Nord Gilaj
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA; Department of Chemistry, Lehman College, Bronx, New York, USA
| | - Karina Peregrina
- Department of Oncology, Albert Einstein College of Medicine, Bronx, New York, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Isabella Brew
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Elena Tosti
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA; Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Karl Shaffer
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Peter C Tyler
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Winfried Edelmann
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA; Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Leonard H Augenlicht
- Department of Oncology, Albert Einstein College of Medicine, Bronx, New York, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Vern L Schramm
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA.
| |
Collapse
|
3
|
Cooper AJL, Dorai T, Pinto JT, Denton TT. Metabolic Heterogeneity, Plasticity, and Adaptation to "Glutamine Addiction" in Cancer Cells: The Role of Glutaminase and the GTωA [Glutamine Transaminase-ω-Amidase (Glutaminase II)] Pathway. BIOLOGY 2023; 12:1131. [PMID: 37627015 PMCID: PMC10452834 DOI: 10.3390/biology12081131] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/06/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023]
Abstract
Many cancers utilize l-glutamine as a major energy source. Often cited in the literature as "l-glutamine addiction", this well-characterized pathway involves hydrolysis of l-glutamine by a glutaminase to l-glutamate, followed by oxidative deamination, or transamination, to α-ketoglutarate, which enters the tricarboxylic acid cycle. However, mammalian tissues/cancers possess a rarely mentioned, alternative pathway (the glutaminase II pathway): l-glutamine is transaminated to α-ketoglutaramate (KGM), followed by ω-amidase (ωA)-catalyzed hydrolysis of KGM to α-ketoglutarate. The name glutaminase II may be confused with the glutaminase 2 (GLS2) isozyme. Thus, we recently renamed the glutaminase II pathway the "glutamine transaminase-ω-amidase (GTωA)" pathway. Herein, we summarize the metabolic importance of the GTωA pathway, including its role in closing the methionine salvage pathway, and as a source of anaplerotic α-ketoglutarate. An advantage of the GTωA pathway is that there is no net change in redox status, permitting α-ketoglutarate production during hypoxia, diminishing cellular energy demands. We suggest that the ability to coordinate control of both pathways bestows a metabolic advantage to cancer cells. Finally, we discuss possible benefits of GTωA pathway inhibitors, not only as aids to studying the normal biological roles of the pathway but also as possible useful anticancer agents.
Collapse
Affiliation(s)
- Arthur J. L. Cooper
- Department of Biochemistry and Molecular Biology, New York Medical College, 15 Dana Road, Valhalla, NY 10595, USA; (T.D.); (J.T.P.)
| | - Thambi Dorai
- Department of Biochemistry and Molecular Biology, New York Medical College, 15 Dana Road, Valhalla, NY 10595, USA; (T.D.); (J.T.P.)
- Department of Urology, New York Medical College, Valhalla, NY 10595, USA
| | - John T. Pinto
- Department of Biochemistry and Molecular Biology, New York Medical College, 15 Dana Road, Valhalla, NY 10595, USA; (T.D.); (J.T.P.)
| | - Travis T. Denton
- Department Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, Washington State University Health Sciences Spokane, Spokane, WA 99202, USA
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University Health Sciences Spokane, Spokane, WA 99164, USA
- Steve Gleason Institute for Neuroscience, Washington State University Health Sciences Spokane, Spokane, WA 99164, USA
| |
Collapse
|
4
|
Salita T, Rustam YH, Hofferek V, Jackson M, Tollestrup I, Sheridan JP, Schramm VL, Evans GB, Reid GE, Munkacsi AB. Phosphoinositide and redox dysregulation by the anticancer methylthioadenosine phosphorylase transition state inhibitor. Biochim Biophys Acta Mol Cell Biol Lipids 2023:159346. [PMID: 37301365 DOI: 10.1016/j.bbalip.2023.159346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/05/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023]
Abstract
Methylthio-DADMe-immucillin-A (MTDIA) is an 86 picomolar inhibitor of 5'-methylthioadenosine phosphorylase (MTAP) with potent and specific anti-cancer efficacy. MTAP salvages S-adenosylmethionine (SAM) from 5'-methylthioadenosine (MTA), a toxic metabolite produced during polyamine biosynthesis. Changes in MTAP expression are implicated in cancer growth and development, making MTAP an appealing target for anti-cancer therapeutics. Since SAM is involved in lipid metabolism, we hypothesised that MTDIA alters the lipidomes of MTDIA-treated cells. To identify these effects, we analysed the lipid profiles of MTDIA-treated Saccharomyces cerevisiae using ultra-high resolution accurate mass spectrometry (UHRAMS). MTAP inhibition by MTDIA, and knockout of the Meu1 gene that encodes for MTAP in yeast, caused global lipidomic changes and differential abundance of lipids involved in cell signaling. The phosphoinositide kinase/phosphatase signaling network was specifically impaired upon MTDIA treatment, and was independently validated and further characterised via altered localization of proteins integral to this network. Functional consequences of dysregulated lipid metabolism included a decrease in reactive oxygen species (ROS) levels induced by MTDIA that was contemporaneous with changes in immunological response factors (nitric oxide, tumour necrosis factor-alpha and interleukin-10) in mammalian cells. These results indicate that lipid homeostasis alterations and concomitant downstream effects may be associated with MTDIA mechanistic efficacy.
Collapse
Affiliation(s)
- Timothy Salita
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand; Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Australia
| | - Yepy H Rustam
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Australia
| | - Vinzenz Hofferek
- School of Chemistry, University of Melbourne, Parkville, Australia
| | - Michael Jackson
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Isaac Tollestrup
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Jeffrey P Sheridan
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Vern L Schramm
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Gary B Evans
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Gavin E Reid
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Australia; School of Chemistry, University of Melbourne, Parkville, Australia; Bio21 Molecular Science & Biotechnology Institute, University of Melbourne, Parkville, Australia
| | - Andrew B Munkacsi
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand.
| |
Collapse
|
5
|
Berg JA, Zhou Y, Ouyang Y, Cluntun AA, Waller TC, Conway ME, Nowinski SM, Van Ry T, George I, Cox JE, Wang B, Rutter J. Metaboverse enables automated discovery and visualization of diverse metabolic regulatory patterns. Nat Cell Biol 2023; 25:616-625. [PMID: 37012464 PMCID: PMC10104781 DOI: 10.1038/s41556-023-01117-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 02/24/2023] [Indexed: 04/05/2023]
Abstract
Metabolism is intertwined with various cellular processes, including controlling cell fate, influencing tumorigenesis, participating in stress responses and more. Metabolism is a complex, interdependent network, and local perturbations can have indirect effects that are pervasive across the metabolic network. Current analytical and technical limitations have long created a bottleneck in metabolic data interpretation. To address these shortcomings, we developed Metaboverse, a user-friendly tool to facilitate data exploration and hypothesis generation. Here we introduce algorithms that leverage the metabolic network to extract complex reaction patterns from data. To minimize the impact of missing measurements within the network, we introduce methods that enable pattern recognition across multiple reactions. Using Metaboverse, we identify a previously undescribed metabolite signature that correlated with survival outcomes in early stage lung adenocarcinoma patients. Using a yeast model, we identify metabolic responses suggesting an adaptive role of citrate homeostasis during mitochondrial dysfunction facilitated by the citrate transporter, Ctp1. We demonstrate that Metaboverse augments the user's ability to extract meaningful patterns from multi-omics datasets to develop actionable hypotheses.
Collapse
Affiliation(s)
- Jordan A Berg
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA.
- Altos Labs, Redwood City, CA, USA.
| | - Youjia Zhou
- School of Computing, University of Utah, Salt Lake City, UT, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA
| | - Yeyun Ouyang
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
- Altos Labs, Redwood City, CA, USA
| | - Ahmad A Cluntun
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - T Cameron Waller
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Megan E Conway
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Sara M Nowinski
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Tyler Van Ry
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
- Metabolomics Core Facility, University of Utah, Salt Lake City, UT, USA
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Ian George
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - James E Cox
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
- Metabolomics Core Facility, University of Utah, Salt Lake City, UT, USA
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
| | - Bei Wang
- School of Computing, University of Utah, Salt Lake City, UT, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA
| | - Jared Rutter
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA.
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA.
- Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
6
|
Brough D, Amos H, Turley K, Murkin J. Trends in Subcutaneous Tumour Height and Impact on Measurement Accuracy. Cancer Inform 2023; 22:11769351231165181. [PMID: 37113645 PMCID: PMC10126793 DOI: 10.1177/11769351231165181] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/05/2023] [Indexed: 04/29/2023] Open
Abstract
Tumour volume is typically calculated using only length and width measurements, using width as a proxy for height in a 1:1 ratio. When tracking tumour growth over time, important morphological information and measurement accuracy is lost by ignoring height, which we show is a unique variable. Lengths, widths, and heights of 9522 subcutaneous tumours in mice were measured using 3D and thermal imaging. The average height:width ratio was found to be 1:3 proving that using width as a proxy for height overestimates tumour volume. Comparing volumes calculated with and without tumour height to the true volumes of excised tumours indeed showed that using the volume formula including height produced volumes 36X more accurate (based off of percentage difference). Monitoring the height:width relationship (prominence) across tumour growth curves indicated that prominence varied, and that height could change independent of width. Twelve cell lines were investigated individually; the scale of tumour prominence was cell line-dependent with relatively less prominent tumours (MC38, BL2, LL/2) and more prominent tumours (RENCA, HCT116) detected. Prominence trends across the growth cycle were also dependent on cell line; prominence was correlated with tumour growth in some cell lines (4T1, CT26, LNCaP), but not others (MC38, TC-1, LL/2). When pooled, invasive cell lines produced tumours that were significantly less prominent at volumes >1200 mm3 compared to non-invasive cell lines (P < .001). Modelling was used to show the impact of the increased accuracy gained by including height in volume calculations on several efficacy study outcomes. Variations in measurement accuracy contribute to experimental variation and irreproducibility of data, therefore we strongly advise researchers to measure height to improve accuracy in tumour studies.
Collapse
Affiliation(s)
- Daniel Brough
- Daniel Brough, BioVolume Ltd, Witney Business & Innovation Centre, Windrush Industrial Park, Burford Road, Witney OX29 7DX, UK.
| | | | | | | |
Collapse
|
7
|
Patro CPK, Biswas N, Pingle SC, Lin F, Anekoji M, Jones LD, Kesari S, Wang F, Ashili S. MTAP loss: a possible therapeutic approach for glioblastoma. J Transl Med 2022; 20:620. [PMID: 36572880 PMCID: PMC9791736 DOI: 10.1186/s12967-022-03823-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 12/11/2022] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma is the most lethal form of brain tumor with a recurrence rate of almost 90% and a survival time of only 15 months post-diagnosis. It is a highly heterogeneous, aggressive, and extensively studied tumor. Multiple studies have proposed therapeutic approaches to mitigate or improve the survival for patients with glioblastoma. In this article, we review the loss of the 5'-methylthioadenosine phosphorylase (MTAP) gene as a potential therapeutic approach for treating glioblastoma. MTAP encodes a metabolic enzyme required for the metabolism of polyamines and purines leading to DNA synthesis. Multiple studies have explored the loss of this gene and have shown its relevance as a therapeutic approach to glioblastoma tumor mitigation; however, other studies show that the loss of MTAP does not have a major impact on the course of the disease. This article reviews the contrasting findings of MTAP loss with regard to mitigating the effects of glioblastoma, and also focuses on multiple aspects of MTAP loss in glioblastoma by providing insights into the known findings and some of the unexplored areas of this field where new approaches can be imagined for novel glioblastoma therapeutics.
Collapse
Affiliation(s)
- C. Pawan K. Patro
- CureScience, 5820 Oberlin Dr, 202, San Diego, CA 92121 USA ,grid.4280.e0000 0001 2180 6431Present Address: Cancer Science Institute, National University of Singapore, Singapore, 117599 Singapore
| | | | | | - Feng Lin
- CureScience, 5820 Oberlin Dr, 202, San Diego, CA 92121 USA
| | - Misa Anekoji
- CureScience, 5820 Oberlin Dr, 202, San Diego, CA 92121 USA
| | | | - Santosh Kesari
- grid.416507.10000 0004 0450 0360Department of Translational Neurosciences, Pacific Neuroscience Institute and Saint John’s Cancer Institute at Providence Saint John’s Health Center, CA 90404 Santa Monica, USA
| | - Feng Wang
- grid.412901.f0000 0004 1770 1022Department of Medical Oncology, Cancer Center, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | | |
Collapse
|
8
|
Madapoosi SS, Cruickshank-Quinn C, Opron K, Erb-Downward JR, Begley LA, Li G, Barjaktarevic I, Barr RG, Comellas AP, Couper DJ, Cooper CB, Freeman CM, Han MK, Kaner RJ, Labaki W, Martinez FJ, Ortega VE, Peters SP, Paine R, Woodruff P, Curtis JL, Huffnagle GB, Stringer KA, Bowler RP, Esther CR, Reisdorph N, Huang YJ. Lung Microbiota and Metabolites Collectively Associate with Clinical Outcomes in Milder Stage Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2022; 206:427-439. [PMID: 35536732 PMCID: PMC11418810 DOI: 10.1164/rccm.202110-2241oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022] Open
Abstract
Rationale: Chronic obstructive pulmonary disease (COPD) is variable in its development. Lung microbiota and metabolites collectively may impact COPD pathophysiology, but relationships to clinical outcomes in milder disease are unclear. Objectives: Identify components of the lung microbiome and metabolome collectively associated with clinical markers in milder stage COPD. Methods: We analyzed paired microbiome and metabolomic data previously characterized from bronchoalveolar lavage fluid in 137 participants in the SPIROMICS (Subpopulations and Intermediate Outcome Measures in COPD Study), or (GOLD [Global Initiative for Chronic Obstructive Lung Disease Stage 0-2). Datasets used included 1) bacterial 16S rRNA gene sequencing; 2) untargeted metabolomics of the hydrophobic fraction, largely comprising lipids; and 3) targeted metabolomics for a panel of hydrophilic compounds previously implicated in mucoinflammation. We applied an integrative approach to select features and model 14 individual clinical variables representative of known associations with COPD trajectory (lung function, symptoms, and exacerbations). Measurements and Main Results: The majority of clinical measures associated with the lung microbiome and metabolome collectively in overall models (classification accuracies, >50%, P < 0.05 vs. chance). Lower lung function, COPD diagnosis, and greater symptoms associated positively with Streptococcus, Neisseria, and Veillonella, together with compounds from several classes (glycosphingolipids, glycerophospholipids, polyamines and xanthine, an adenosine metabolite). In contrast, several Prevotella members, together with adenosine, 5'-methylthioadenosine, sialic acid, tyrosine, and glutathione, associated with better lung function, absence of COPD, or less symptoms. Significant correlations were observed between specific metabolites and bacteria (Padj < 0.05). Conclusions: Components of the lung microbiome and metabolome in combination relate to outcome measures in milder COPD, highlighting their potential collaborative roles in disease pathogenesis.
Collapse
Affiliation(s)
| | | | - Kristopher Opron
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | | | - Lesa A. Begley
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Gen Li
- Department of Biostatistics, School of Public Health
| | | | - R. Graham Barr
- Department of Medicine and
- Department of Epidemiology, Columbia University Medical Center, New York, New York
| | | | | | | | | | - MeiLan K. Han
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | | | - Wassim Labaki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | | | - Victor E. Ortega
- Wake Forest School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Stephen P. Peters
- Wake Forest School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | | | - Prescott Woodruff
- University of California at San Francisco, San Francisco, California
| | - Jeffrey L. Curtis
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Gary B. Huffnagle
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Molecular, Cellular and Developmental Biology
| | | | - Russell P. Bowler
- School of Medicine, University of Colorado, Aurora, Colorado; and
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Charles R. Esther
- Division of Pediatric Pulmonology, and
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Campus, Aurora, Colorado
| | - Yvonne J. Huang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
9
|
Du C, Li SW, Singh SX, Roso K, Sun MA, Pirozzi CJ, Yang R, Li JL, He Y. Epigenetic Regulation of Fanconi Anemia Genes Implicates PRMT5 Blockage as a Strategy for Tumor Chemosensitization. Mol Cancer Res 2021; 19:2046-2056. [PMID: 34521764 DOI: 10.1158/1541-7786.mcr-21-0093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/16/2021] [Accepted: 08/31/2021] [Indexed: 11/16/2022]
Abstract
Strengthened DNA repair pathways in tumor cells contribute to the development of resistance to DNA-damaging agents. Consequently, targeting proteins in these pathways is a promising strategy for tumor chemosensitization. Here, we show that the expression of a subset of Fanconi anemia (FA) genes is attenuated in glioblastoma tumor cells deficient in methylthioadenosine phosphorylase (MTAP), a common genetic alteration in a variety of cancers. Subsequent experiments in cell line models of different cancer types illustrate that this reduced transcription of FA genes can be recapitulated by blockage of Protein Arginine Methyltransferase 5 (PRMT5), a promising therapeutically targetable epigenetic regulator whose enzymatic activity is compromised in MTAP-deficient cells. Further analyses provide evidence to support that PRMT5 can function as an epigenetic regulator that contributes to the increased expression of FA genes in cancer cells. Most notably and consistent with the essential roles of FA proteins in resolving DNA damage elicited by interstrand crosslinking (ICL) agents, PRMT5 blockage, as well as MTAP loss, sensitizes tumor cells to ICL agents both in vitro and in xenografts. Collectively, these findings reveal a novel epigenetic mechanism underlying the upregulated expression of FA genes in cancer cells and suggest that therapeutically targeting PRMT5 can have an additional benefit of chemosensitizing tumor cells to ICL agents. IMPLICATIONS: PRMT5 positively regulates the expression of FA genes. Inhibition of PRMT5 attenuates FA-dependent DNA repair pathway and sensitizes tumor cells to ICL agents.
Collapse
Affiliation(s)
- Changzheng Du
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina.,Department of Pathology, Duke University Medical Center, Durham, North Carolina.,School of Medicine, Southern University of Science and Technology, and Southern University of Science and Technology Hospital, Nanshan District, Shenzhen, Guangdong, China
| | - Steven W Li
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina
| | - Simranjit X Singh
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina.,Department of Pathology, Duke University Medical Center, Durham, North Carolina.,Pathology Graduate Program, Duke University Medical Center, Durham, North Carolina
| | - Kristen Roso
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina.,Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Michael A Sun
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina.,Department of Pathology, Duke University Medical Center, Durham, North Carolina.,Pathology Graduate Program, Duke University Medical Center, Durham, North Carolina
| | - Christopher J Pirozzi
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina.,Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Rui Yang
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina.,Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Jian-Liang Li
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, Durham, North Carolina
| | - Yiping He
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina. .,Department of Pathology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
10
|
Firestone RS, Feng M, Basu I, Peregrina K, Augenlicht LH, Schramm VL. Transition state analogue of MTAP extends lifespan of APC Min/+ mice. Sci Rep 2021; 11:8844. [PMID: 33893330 PMCID: PMC8065027 DOI: 10.1038/s41598-021-87734-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 03/23/2021] [Indexed: 01/03/2023] Open
Abstract
A mouse model of human Familial Adenomatous Polyposis responds favorably to pharmacological inhibition of 5'-methylthioadenosine phosphorylase (MTAP). Methylthio-DADMe-Immucillin-A (MTDIA) is an orally available, transition state analogue inhibitor of MTAP. 5'-Methylthioadenosine (MTA), the substrate for MTAP, is formed in polyamine synthesis and is recycled by MTAP to S-adenosyl-L-methionine (SAM) via salvage pathways. MTDIA treatment causes accumulation of MTA, which inhibits growth of human head and neck (FaDu) and lung (H359, A549) cancers in immunocompromised mouse models. We investigated the efficacy of oral MTDIA as an anti-cancer therapeutic for intestinal adenomas in immunocompetent APCMin/+ mice, a murine model of human Familial Adenomatous Polyposis. Tumors in APCMin/+ mice were decreased in size by MTDIA treatment, resulting in markedly improved anemia and doubling of mouse lifespan. Metabolomic analysis of treated mice showed no changes in polyamine, methionine, SAM or ATP levels when compared with control mice but indicated an increase in MTA, the MTAP substrate. Generation of an MTDIA-resistant cell line in culture showed a four-fold amplification of the methionine adenosyl transferase (MAT2A) locus and expression of this enzyme. MAT2A is downstream of MTAP action and catalyzes synthesis of the SAM necessary for methylation reactions. Immunohistochemical analysis of treated mouse intestinal tissue demonstrated a decrease in symmetric dimethylarginine, a PRMT5-catalyzed modification. The anti-cancer effects of MTDIA indicate that increased cellular MTA inhibits PRMT5-mediated methylations resulting in attenuated tumor growth. Oral dosing of MTDIA as monotherapy has potential for delaying the onset and progression of colorectal cancers in Familial Adenomatous Polyposis (FAP) as well as residual duodenal tumors in FAP patients following colectomy. MTDIA causes a physiologic inactivation of MTAP and may also have efficacy in combination with inhibitors of MAT2A or PRMT5, known synthetic-lethal interactions in MTAP-/- cancer cell lines.
Collapse
Affiliation(s)
- Ross S Firestone
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mu Feng
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Indranil Basu
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Karina Peregrina
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Leonard H Augenlicht
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| | - Vern L Schramm
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
11
|
Liu W, Zhuang C, Huang T, Yang S, Zhang M, Lin B, Jiang Y. Loss of CDKN2A at chromosome 9 has a poor clinical prognosis and promotes lung cancer progression. Mol Genet Genomic Med 2020; 8:e1521. [PMID: 33155773 PMCID: PMC7767555 DOI: 10.1002/mgg3.1521] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/12/2020] [Accepted: 09/03/2020] [Indexed: 11/23/2022] Open
Abstract
Objective This study aimed to identify critical genes involved in the tumor biology of lung cancer via datamining of The Cancer Genome Atlas (TCGA) with special focus on gene copy number variation. Methods Genomic deletion and amplification were analyzed with cBioportal online tools. Relative expression of Cyclin Dependent Kinase Inhibitor 2A (CDKN2A) was analyzed by both real‐time polymerase chain reaction (PCR) and Western blot. The abundance of methylthioadenosine phosphorylase (MTAP) and epithelial‐mesenchymal transition markers were analyzed by real‐time PCR. Cell proliferation was determined by cell counting kit‐8 method and cell viability was measured with 3‐(4,5‐dimethylthiazol‐2‐yl)‐2,5‐diphenyltetrazolium bromide assay. The cell migration and invasion were measured with transwell chamber assay, and migrative capacity was further evaluated by wound healing assay. Results We found the frequent loss of CDKN2A was associated with its downregulation in lung cancer, and siRNA‐mediated CDNKN2A knockdown significantly stimulated cell proliferation, invasion, and migration. Mechanistically, we unraveled that MTAP, which was positively correlated with CDKN2A, predominantly mediated the antitumoral function of CDKN2A in lung cancer. Conclusion Our study consolidated the involvement of CDKN2A‐MTAP signaling in the context of lung cancer.
Collapse
Affiliation(s)
- Wei Liu
- Department of Respiratory and Critical Care Medicine, The 900th Hospital of Joint Logistic Support Force, Fuzhou, Fujian, China
| | - Congwen Zhuang
- Department of Respiratory and Critical Care Medicine, The 900th Hospital of Joint Logistic Support Force, Fuzhou, Fujian, China
| | - Tengfei Huang
- Department of Respiratory and Critical Care Medicine, The 900th Hospital of Joint Logistic Support Force, Fuzhou, Fujian, China
| | - Shengsheng Yang
- Department of Respiratory and Critical Care Medicine, The 900th Hospital of Joint Logistic Support Force, Fuzhou, Fujian, China
| | - Meiqing Zhang
- Department of Respiratory and Critical Care Medicine, The 900th Hospital of Joint Logistic Support Force, Fuzhou, Fujian, China
| | - Baoquan Lin
- Department of Thoracic Surgery, The 900th Hospital of Joint Logistic Support Force, Fuzhou, Fujian, China
| | - Yi Jiang
- Department of Hepatobiliary Surgery, The 900th Hospital of Joint Logistic Support Force, Fuzhou, Fujian, China
| |
Collapse
|
12
|
Du C, Hansen LJ, Singh SX, Wang F, Sun R, Moure CJ, Roso K, Greer PK, Yan H, He Y. A PRMT5-RNF168-SMURF2 Axis Controls H2AX Proteostasis. Cell Rep 2020; 28:3199-3211.e5. [PMID: 31533041 DOI: 10.1016/j.celrep.2019.08.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 06/11/2019] [Accepted: 08/06/2019] [Indexed: 12/21/2022] Open
Abstract
H2AX safeguards genomic stability in a dose-dependent manner; however, mechanisms governing its proteostasis are poorly understood. Here, we identify a PRMT5-RNF168-SMURF2 cascade that regulates H2AX proteostasis. We show that PRMT5 sustains the expression of RNF168, an E3 ubiquitin ligase essential for DNA damage response (DDR). Suppression of PRMT5 occurs in methylthioadenosine phosphorylase (MTAP)-deficient glioblastoma cells and attenuates the expression of RNF168, leading to destabilization of H2AX by E3 ubiquitin ligase SMURF2. RNF168 and SMURF2 serve as a stabilizer and destabilizer of H2AX, respectively, via their dynamic interactions with H2AX. In supporting an important role of this signaling cascade in regulating H2AX, MTAP-deficient glioblastoma cells display higher levels of DNA damage spontaneously or in response to genotoxic agents. These findings reveal a regulatory mechanism of H2AX proteostasis and define a signaling cascade that is essential to DDR and that is disrupted by the loss of a metabolic enzyme in tumor cells.
Collapse
Affiliation(s)
- Changzheng Du
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA; Gastrointestinal Cancer Center, Peking University Cancer Hospital, Beijing 100142, China
| | - Landon J Hansen
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Simranjit X Singh
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA; Pathology Graduate Program, Duke University Medical Center, Durham, NC, USA
| | - Feiyifan Wang
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC 27710, USA
| | - Ran Sun
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA; Scientific Research Center, China-Japan Union Hospital, Jilin University, Jilin 130033, China
| | - Casey J Moure
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Kristen Roso
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Paula K Greer
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Hai Yan
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yiping He
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
13
|
Loss of 5'-Methylthioadenosine Phosphorylase (MTAP) is Frequent in High-Grade Gliomas; Nevertheless, it is Not Associated with Higher Tumor Aggressiveness. Cells 2020; 9:cells9020492. [PMID: 32093414 PMCID: PMC7072758 DOI: 10.3390/cells9020492] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/04/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
The 5’-methylthioadenosine phosphorylase (MTAP) gene is located in the chromosomal region 9p21. MTAP deletion is a frequent event in a wide variety of human cancers; however, its biological role in tumorigenesis remains unclear. The purpose of this study was to characterize the MTAP expression profile in a series of gliomas and to associate it with patients’ clinicopathological features. Moreover, we sought to evaluate, through glioma gene-edited cell lines, the biological impact of MTAP in gliomas. MTAP expression was evaluated in 507 glioma patients by immunohistochemistry (IHC), and the expression levels were associated with patients’ clinicopathological features. Furthermore, an in silico study was undertaken using genomic databases totalizing 350 samples. In glioma cell lines, MTAP was edited, and following MTAP overexpression and knockout (KO), a transcriptome analysis was performed by NanoString Pan-Cancer Pathways panel. Moreover, MTAP’s role in glioma cell proliferation, migration, and invasion was evaluated. Homozygous deletion of 9p21 locus was associated with a reduction of MTAP mRNA expression in the TCGA (The Cancer Genome Atlas) - glioblastoma dataset (p < 0.01). In addition, the loss of MTAP expression was markedly high in high-grade gliomas (46.6% of cases) determined by IHC and Western blotting (40% of evaluated cell lines). Reduced MTAP expression was associated with a better prognostic in the adult glioblastoma dataset (p < 0.001). Nine genes associated with five pathways were differentially expressed in MTAP-knockout (KO) cells, with six upregulated and three downregulated in MTAP. Analysis of cell proliferation, migration, and invasion did not show any significant differences between MTAP gene-edited and control cells. Our results integrating data from patients as well as in silico and in vitro models provide evidence towards the lack of strong biological importance of MTAP in gliomas. Despite the frequent loss of MTAP, it seems not to have a clinical impact in survival and does not act as a canonic tumor suppressor gene in gliomas.
Collapse
|
14
|
MTAP-deficiency could predict better treatment response in advanced lung adenocarcinoma patients initially treated with pemetrexed-platinum chemotherapy and bevacizumab. Sci Rep 2020; 10:843. [PMID: 31965001 PMCID: PMC6972892 DOI: 10.1038/s41598-020-57812-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 01/08/2020] [Indexed: 12/22/2022] Open
Abstract
To investigate the predictive value of methylthioadenosine phosphorylase (MTAP) on treatment response and survival in advanced lung adenocarcinoma. MTAP expression was detected by immunohistochemistry. Treatment response and survival were compared according to MTAP expression level. The results indicated MTAP-low expression was observed in 61.2% (101/165) of all patients. The objective response rate and disease control rate improved in the MTAP-low group (64.4% vs 46.9%, p = 0.035; 92.1% vs. 79.7%, p = 0.03; respectively). The median progression-free survival and survival time in the MTAP-low group were significantly lower than that in the MTAP-high group (8.1 vs. 13.1 months, p = 0.002; 22 vs. 32 months, p = 0.044). Multivariate analysis demonstrated that brain metastasis (HR 1.55, p = 0.046), thoracic radiation (HR 0.52, p = 0.026), and MTAP-low expression (HR 1.36, p = 0.038) were independent factors on survival. It is concluded that MTAP-low expression could predict improved treatment response but worsened survival in advanced lung adenocarcinoma.
Collapse
|
15
|
Pharmacological polyamine catabolism upregulation with methionine salvage pathway inhibition as an effective prostate cancer therapy. Nat Commun 2020; 11:52. [PMID: 31911608 PMCID: PMC6946658 DOI: 10.1038/s41467-019-13950-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 12/06/2019] [Indexed: 01/16/2023] Open
Abstract
Prostatic luminal epithelial cells secrete high levels of acetylated polyamines into the prostatic lumen, sensitizing them to perturbations of connected metabolic pathways. Enhanced flux is driven by spermidine/spermine N1-acetyltransferase (SSAT) activity, which acetylates polyamines leading to their secretion and drives biosynthetic demand. The methionine salvage pathway recycles one-carbon units lost to polyamine biosynthesis to the methionine cycle to overcome stress. Prostate cancer (CaP) relies on methylthioadenosine phosphorylase (MTAP), the rate-limiting enzyme, to relieve strain. Here, we show that inhibition of MTAP alongside SSAT upregulation is synergistic in androgen sensitive and castration recurrent CaP models in vitro and in vivo. The combination treatment increases apoptosis in radical prostatectomy ex vivo explant samples. This unique high metabolic flux through polyamine biosynthesis and connected one carbon metabolism in CaP creates a metabolic dependency. Enhancing this flux while simultaneously targeting this dependency in prostate cancer results in an effective therapeutic approach potentially translatable to the clinic. Prostate cancer cells depend on MTAP, the rate-limiting enzyme involved in the methionine salvage pathway, to cope with increased polyamine biosynthesis. Here, the authors show that inducing upregulation of polyamine biosynthesis and targeting MTAP synergize to increase apoptosis in prostate cancer cells.
Collapse
|
16
|
Harijan RK, Hoff O, Ducati RG, Firestone RS, Hirsch BM, Evans GB, Schramm VL, Tyler PC. Selective Inhibitors of Helicobacter pylori Methylthioadenosine Nucleosidase and Human Methylthioadenosine Phosphorylase. J Med Chem 2019; 62:3286-3296. [PMID: 30860833 PMCID: PMC6635953 DOI: 10.1021/acs.jmedchem.8b01642] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Bacterial 5'-methylthioadenosine/ S-adenosylhomocysteine nucleosidase (MTAN) hydrolyzes adenine from its substrates to form S-methyl-5-thioribose and S-ribosyl-l-homocysteine. MTANs are involved in quorum sensing, menaquinone synthesis, and 5'-methylthioadenosine recycling to S-adenosylmethionine. Helicobacter pylori uses MTAN in its unusual menaquinone pathway, making H. pylori MTAN a target for antibiotic development. Human 5'-methylthioadenosine phosphorylase (MTAP), a reported anticancer target, catalyzes phosphorolysis of 5'-methylthioadenosine to salvage S-adenosylmethionine. Transition-state analogues designed for HpMTAN and MTAP show significant overlap in specificity. Fifteen unique transition-state analogues are described here and are used to explore inhibitor specificity. Several analogues of HpMTAN bind in the picomolar range while inhibiting human MTAP with orders of magnitude weaker affinity. Structural analysis of HpMTAN shows inhibitors extending through a hydrophobic channel to the protein surface. The more enclosed catalytic sites of human MTAP require the inhibitors to adopt a folded structure, displacing the phosphate nucleophile from the catalytic site.
Collapse
Affiliation(s)
- Rajesh K. Harijan
- Department of Biochemistry, Albert Einstein College
of Medicine, New York 10461, New York, United States
| | - Oskar Hoff
- Ferrier Research Institute, Victoria University of
Wellington, Wellington 5040, New Zealand
| | - Rodrigo G. Ducati
- Department of Biochemistry, Albert Einstein College
of Medicine, New York 10461, New York, United States
| | - Ross S. Firestone
- Department of Biochemistry, Albert Einstein College
of Medicine, New York 10461, New York, United States
| | - Brett M. Hirsch
- Department of Biochemistry, Albert Einstein College
of Medicine, New York 10461, New York, United States
| | - Gary B. Evans
- Ferrier Research Institute, Victoria University of
Wellington, Wellington 5040, New Zealand
| | - Vern L. Schramm
- Department of Biochemistry, Albert Einstein College
of Medicine, New York 10461, New York, United States
| | - Peter C. Tyler
- Ferrier Research Institute, Victoria University of
Wellington, Wellington 5040, New Zealand
| |
Collapse
|
17
|
Polineni D, Dang H, Gallins PJ, Jones LC, Pace RG, Stonebraker JR, Commander LA, Krenicky JE, Zhou YH, Corvol H, Cutting GR, Drumm ML, Strug LJ, Boyle MP, Durie PR, Chmiel JF, Zou F, Wright FA, O'Neal WK, Knowles MR. Airway Mucosal Host Defense Is Key to Genomic Regulation of Cystic Fibrosis Lung Disease Severity. Am J Respir Crit Care Med 2019; 197:79-93. [PMID: 28853905 DOI: 10.1164/rccm.201701-0134oc] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
RATIONALE The severity of cystic fibrosis (CF) lung disease varies widely, even for Phe508del homozygotes. Heritability studies show that more than 50% of the variability reflects non-cystic fibrosis transmembrane conductance regulator (CFTR) genetic variation; however, the full extent of the pertinent genetic variation is not known. OBJECTIVES We sought to identify novel CF disease-modifying mechanisms using an integrated approach based on analyzing "in vivo" CF airway epithelial gene expression complemented with genome-wide association study (GWAS) data. METHODS Nasal mucosal RNA from 134 patients with CF was used for RNA sequencing. We tested for associations of transcriptomic (gene expression) data with a quantitative phenotype of CF lung disease severity. Pathway analysis of CF GWAS data (n = 5,659 patients) was performed to identify novel pathways and assess the concordance of genomic and transcriptomic data. Association of gene expression with previously identified CF GWAS risk alleles was also tested. MEASUREMENTS AND MAIN RESULTS Significant evidence of heritable gene expression was identified. Gene expression pathways relevant to airway mucosal host defense were significantly associated with CF lung disease severity, including viral infection, inflammation/inflammatory signaling, lipid metabolism, apoptosis, ion transport, Phe508del CFTR processing, and innate immune responses, including HLA (human leukocyte antigen) genes. Ion transport and CFTR processing pathways, as well as HLA genes, were identified across differential gene expression and GWAS signals. CONCLUSIONS Transcriptomic analyses of CF airway epithelia, coupled to genomic (GWAS) analyses, highlight the role of heritable host defense variation in determining the pathophysiology of CF lung disease. The identification of these pathways provides opportunities to pursue targeted interventions to improve CF lung health.
Collapse
Affiliation(s)
- Deepika Polineni
- 1 Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas.,2 Cystic Fibrosis/Pulmonary Research and Treatment Center, Marsico Lung Institute, School of Medicine, and
| | - Hong Dang
- 2 Cystic Fibrosis/Pulmonary Research and Treatment Center, Marsico Lung Institute, School of Medicine, and
| | - Paul J Gallins
- 3 Bioinformatics Research Center, Department of Biological Sciences
| | - Lisa C Jones
- 2 Cystic Fibrosis/Pulmonary Research and Treatment Center, Marsico Lung Institute, School of Medicine, and
| | - Rhonda G Pace
- 2 Cystic Fibrosis/Pulmonary Research and Treatment Center, Marsico Lung Institute, School of Medicine, and
| | - Jaclyn R Stonebraker
- 2 Cystic Fibrosis/Pulmonary Research and Treatment Center, Marsico Lung Institute, School of Medicine, and
| | - Leah A Commander
- 2 Cystic Fibrosis/Pulmonary Research and Treatment Center, Marsico Lung Institute, School of Medicine, and
| | - Jeanne E Krenicky
- 4 Department of Pediatrics, Rainbow Babies and Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Yi-Hui Zhou
- 3 Bioinformatics Research Center, Department of Biological Sciences
| | - Harriet Corvol
- 5 Pediatric Pulmonary Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Trousseau, Institut National de la Santé et la Recherche Médicale (INSERM) U938, Paris, France.,6 Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Paris 6, Paris, France
| | - Garry R Cutting
- 7 McKusick-Nathans Institute of Genetic Medicine.,8 Department of Pediatrics, and
| | - Mitchell L Drumm
- 4 Department of Pediatrics, Rainbow Babies and Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Lisa J Strug
- 9 Program in Genetics and Genome Biology.,10 Division of Biostatistics, Dalla Lana School of Public Health, and
| | - Michael P Boyle
- 11 Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Peter R Durie
- 12 Physiology and Experimental Medicine Research Program, and.,13 Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, Ontario, Canada; and.,14 Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - James F Chmiel
- 4 Department of Pediatrics, Rainbow Babies and Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Fei Zou
- 15 Department of Biostatistics, School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Fred A Wright
- 16 Department of Statistics, and.,17 Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina
| | - Wanda K O'Neal
- 2 Cystic Fibrosis/Pulmonary Research and Treatment Center, Marsico Lung Institute, School of Medicine, and
| | - Michael R Knowles
- 2 Cystic Fibrosis/Pulmonary Research and Treatment Center, Marsico Lung Institute, School of Medicine, and
| |
Collapse
|
18
|
Li Y, Wang Y, Wu P. 5'-Methylthioadenosine and Cancer: old molecules, new understanding. J Cancer 2019; 10:927-936. [PMID: 30854099 PMCID: PMC6400808 DOI: 10.7150/jca.27160] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 12/17/2018] [Indexed: 12/19/2022] Open
Abstract
While the metabolic changes in cancer tissues were first observed by Warburg Otto almost a century ago, altered metabolism has recently returned as a focus of cancer research. 5'-Methylthioadenosine (MTA) is a naturally occurring sulfur-containing nucleoside found in numerous species. While MTA was first isolated several decades ago, a lack of sensitive and specific analytical methodologies designed for its direct quantification has hampered the study of its physiological and pathophysiological features. Many studies indicate that MTA suppresses tumors by inhibiting tumor cell proliferation, invasion, and the induction of apoptosis while controlling the inflammatory micro-environments of tumor tissue. In this review, we assessed the effects of MTA and of related materials on the growth and functions of normal and malignant cells.
Collapse
Affiliation(s)
- Yaofeng Li
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yubo Wang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Ping Wu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
19
|
Targeting the insulin-like growth factor-1 receptor in MTAP-deficient renal cell carcinoma. Signal Transduct Target Ther 2019; 4:2. [PMID: 30701095 PMCID: PMC6345872 DOI: 10.1038/s41392-019-0035-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 02/06/2023] Open
Abstract
Renal cell carcinoma (RCC) has emerged as a metabolic disease characterized by dysregulated expression of metabolic enzymes. Patients with metastatic RCC have an unusually poor prognosis and near-universal resistance to all current therapies. To improve RCC treatment and the survival rate of patients with RCC, there is an urgent need to reveal the mechanisms by which metabolic reprogramming regulates aberrant signaling and oncogenic progression. Through an integrated analysis of RCC metabolic pathways, we showed that methylthioadenosine phosphorylase (MTAP) and its substrate methylthioadenosine (MTA) are dysregulated in aggressive RCC. A decrease in MTAP expression was observed in RCC tissues and correlated with higher tumor grade and shorter overall survival. Genetic manipulation of MTAP demonstrated that MTAP expression inhibits the epithelial-mesenchymal transition, invasion and migration of RCC cells. Interestingly, we found a decrease in the protein methylation level with a concomitant increase in tyrosine phosphorylation after MTAP knockout. A phospho-kinase array screen identified the type 1 insulin-like growth factor-1 receptor (IGF1R) as the candidate with the highest upregulation in tyrosine phosphorylation in response to MTAP loss. We further demonstrated that IGF1R phosphorylation acts upstream of Src and STAT3 signaling in MTAP-knockout RCC cells. IGF1R suppression by a selective inhibitor of IGF1R, linsitinib, impaired the cell migration and invasion capability of MTAP-deleted cells. Surprisingly, an increase in linsitinib-mediated cytotoxicity occurred in RCC cells with MTAP deficiency. Our data suggest that IGF1R signaling is a driver pathway that contributes to the aggressive nature of MTAP-deleted RCC. A receptor that is triggered by an enzyme deficiency in kidney cancer could act as an anticancer drug target. Ching-Hsien Chen of the University of California Davis and colleagues in the USA and Taiwan found that renal cell carcinomas are deficient in the enzyme methylthioadenosine phosphorylase (MTAP). This deficiency, which correlates with higher tumour grade and shorter overall survival, leads to the activation of type 1 insulin-like growth factor-1 receptor (IGF1R). This in turn activates signaling pathways that support cancer cell survival, growth, and invasiveness. The team found that a selective IGF1R inhibitor, called linsitinib, suppressed colony-forming ability and reduced cell motility in renal carcinoma cells. The findings suggest that IGF1R signaling drives pathways that contribute to the aggressive nature of renal carcinoma cells lacking MTAP.
Collapse
|
20
|
Abstract
Transition state theory teaches that chemically stable mimics of enzymatic transition states will bind tightly to their cognate enzymes. Kinetic isotope effects combined with computational quantum chemistry provides enzymatic transition state information with sufficient fidelity to design transition state analogues. Examples are selected from various stages of drug development to demonstrate the application of transition state theory, inhibitor design, physicochemical characterization of transition state analogues, and their progress in drug development.
Collapse
Affiliation(s)
- Vern L. Schramm
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| |
Collapse
|
21
|
Abstract
Advances in our understanding of the metabolism and molecular functions of polyamines and their alterations in cancer have led to resurgence in the interest of targeting polyamine metabolism as an anticancer strategy. Increasing knowledge of the interplay between polyamine metabolism and other cancer-driving pathways, including the PTEN-PI3K-mTOR complex 1 (mTORC1), WNT signalling and RAS pathways, suggests potential combination therapies that will have considerable clinical promise. Additionally, an expanding number of promising clinical trials with agents targeting polyamines for both therapy and prevention are ongoing. New insights into molecular mechanisms linking dysregulated polyamine catabolism and carcinogenesis suggest additional strategies that can be used for cancer prevention in at-risk individuals. In addition, polyamine blocking therapy, a strategy that combines the inhibition of polyamine biosynthesis with the simultaneous blockade of polyamine transport, can be more effective than therapies based on polyamine depletion alone and may involve an antitumour immune response. These findings open up new avenues of research into exploiting aberrant polyamine metabolism for anticancer therapy.
Collapse
Affiliation(s)
- Robert A Casero
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.
| | - Tracy Murray Stewart
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Anthony E Pegg
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
22
|
Gerner EW, Bruckheimer E, Cohen A. Cancer pharmacoprevention: Targeting polyamine metabolism to manage risk factors for colon cancer. J Biol Chem 2018; 293:18770-18778. [PMID: 30355737 DOI: 10.1074/jbc.tm118.003343] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cancer is a set of diseases characterized by uncontrolled cell growth. In certain cancers of the gastrointestinal tract, the adenomatous polyposis coli (APC) tumor suppressor gene is altered in either germline or somatic cells and causes formation of risk factors, such as benign colonic or intestinal neoplasia, which can progress to invasive cancer. APC is a key component of the WNT pathway, contributing to normal GI tract development, and APC alteration results in dysregulation of the pathway for production of polyamines, which are ubiquitous cations essential for cell growth. Studies with mice have identified nonsteroidal anti-inflammatory drugs (NSAIDs) and difluoromethylornithine (DFMO), an inhibitor of polyamine synthesis, as potent inhibitors of colon carcinogenesis. Moreover, gene expression profiling has uncovered that NSAIDs activate polyamine catabolism and export. Several DFMO-NSAID combination strategies are effective and safe methods for reducing risk factors in clinical trials with patients having genetic or sporadic risk of colon cancer. These strategies affect cancer stem cells, inflammation, immune surveillance, and the microbiome. Pharmacotherapies consisting of drug combinations targeting the polyamine pathway provide a complementary approach to surgery and cytotoxic cancer treatments for treating patients with cancer risk factors. In this Minireview, we discuss the role of polyamines in colon cancer and highlight the mechanisms of select pharmacoprevention agents to delay or prevent carcinogenesis in humans.
Collapse
Affiliation(s)
- Eugene W Gerner
- From Cancer Prevention Pharmaceuticals, Tucson, Arizona 85718 and .,the Department of Cell and Molecular Medicine, University of Arizona, Tucson, Arizona 85711
| | | | - Alfred Cohen
- From Cancer Prevention Pharmaceuticals, Tucson, Arizona 85718 and
| |
Collapse
|
23
|
Methylthioadenosine Suppresses Salmonella Virulence. Infect Immun 2018; 86:IAI.00429-18. [PMID: 29866910 PMCID: PMC6105896 DOI: 10.1128/iai.00429-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 02/01/2023] Open
Abstract
In order to deploy virulence factors at appropriate times and locations, microbes must rapidly sense and respond to various metabolite signals. Previously, we showed a transient elevation of the methionine-derived metabolite methylthioadenosine (MTA) concentration in serum during systemic Salmonella enterica serovar Typhimurium infection. Here we explored the functional consequences of increased MTA concentrations on S Typhimurium virulence. We found that MTA, but not other related metabolites involved in polyamine synthesis and methionine salvage, reduced motility, host cell pyroptosis, and cellular invasion. Further, we developed a genetic model of increased bacterial endogenous MTA production by knocking out the master repressor of the methionine regulon, metJ Like MTA-treated S Typhimurium, the ΔmetJ mutant displayed reduced motility, host cell pyroptosis, and invasion. These phenotypic effects of MTA correlated with suppression of flagellar and Salmonella pathogenicity island 1 (SPI-1) networks. S Typhimurium ΔmetJ had reduced virulence in oral and intraperitoneal infection of C57BL/6J mice independently of the effects of MTA on SPI-1. Finally, ΔmetJ bacteria induced a less severe inflammatory cytokine response in a mouse sepsis model. Together, these data indicate that exposure of S Typhimurium to MTA or disruption of the bacterial methionine metabolism pathway suppresses S Typhimurium virulence.
Collapse
|
24
|
Jaiswal RK, Kumar P, Kumar M, Yadava PK. hTERT promotes tumor progression by enhancing TSPAN13 expression in osteosarcoma cells. Mol Carcinog 2018; 57:1038-1054. [PMID: 29722072 DOI: 10.1002/mc.22824] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/06/2018] [Accepted: 04/14/2018] [Indexed: 01/11/2023]
Abstract
Telomerase complex maintains the length of the telome, cbre, and protects erosion of the physical ends of the eukaryotic chromosome in all actively dividing cells including cancer cells. Telomerase activation extends the lifespan of cells in culture by maintaining the length of the telomere. Compared to terminally differentiated somatic cells, telomerase activity remains high in over 90% of cancer cells. It has now become clear that the role of telomerase is much more complex than just telomere lengthening. The remaining 10% of cancers deploy ALT (alternative lengthening of telomeres) pathway to maintain telomere length. Telomerase inhibitors offer a good therapeutic option. Also, telomerase-associated molecules can be targeted provided their roles are clearly established. In any case, it is necessary to understand the major role of telomerase in cancer cells. Many studies have already been done to explore gene profiling of a telomerase positive cell by knocking down expression of hTERT (telomerase reverse transcriptase). To complement these studies, we performed global gene profiling of a telomerase negative cell by ectopically expressing hTERT and studied changes in the global gene expression patterns. Analysis of microarray data for telomerase negative cells ectopically expressing telomerase showed 76 differentially regulated genes, out of which 39 genes were upregulated, and 37 were downregulated. Three upregulated genes such as TSPAN13, HMGCS2, DLX5, and three downregulated genes like DHRS2, CRYAB, and PDLIM1 were validated by real-time PCR. Knocking down of TSAPN13 in hTERT overexpressing U2OS cells enhanced the apoptosis of the cells. TSPAN13 knockdown in these cells suppressed mesenchymal properties and enhanced epithelial character.
Collapse
Affiliation(s)
- Rishi K Jaiswal
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Pramod Kumar
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.,Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi, India
| | - Manoj Kumar
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Pramod K Yadava
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
25
|
Abstract
The Immucillins are chemically stable analogues that mimic the ribocation and leaving-group features of N-ribosyltransferase transition states. Infectious disease agents often rely on ribosyltransferase chemistry in pathways involving precursor synthesis for nucleic acids, salvage of nucleic acid precursors, or synthetic pathways with nucleoside intermediates. Here, we review three infectious agents and the use of the Immucillins to taget enzymes essential to the parasites. First, DADMe-Immucillin-G is a purine nucleoside phosphorylase (PNP) inhibitor that blocks purine salvage and shows clinical potential for treatment for the malaria parasite Plasmodium falciparum, a purine auxotroph requiring hypoxanthine for purine nucleotide synthesis. Inhibition of the PNPs in the host and in parasite cells leads to apurinic starvation and death. Second, Helicobacter pylori, a causative agent of human ulcers, synthesizes menaquinone, an essential electron transfer agent, in a pathway requiring aminofutalosine nucleoside hydrolysis. Inhibitors of the H. pylori methylthioadenosine nucleosidase (MTAN) are powerful antibiotics for this organism. Synthesis of menaquinone by the aminofutalosine pathway does not occur in most bacteria populating the human gut microbiome. Thus, MTAN inhibitors provide high-specificity antibiotics for H. pylori and are not expected to disrupt the normal gut bacterial flora. Third, Immucillin-A was designed as a transition state analogue of the atypical PNP from Trichomonas vaginalis. In antiviral screens, Immucillin-A was shown to act as a prodrug. It is active against filoviruses and flaviviruses. In virus-infected cells, Immucillin-A is converted to the triphosphate, is incorporated into the viral transcript, and functions as an atypical chain-terminator for RNA-dependent RNA polymerases. Immucillin-A has entered clinical trials for use as an antiviral. We also summarize other Immucillins that have been characterized in successful clinical trials for T-cell lymphoma and gout. The human trials support the potential development of the Immucillins in infectious diseases.
Collapse
Affiliation(s)
- Gary B. Evans
- Ferrier Research
Institute, Victoria University of Wellington, 69 Gracefield Road, Gracefield, Lower Hutt, 5010, New Zealand
| | - Peter C. Tyler
- Ferrier Research
Institute, Victoria University of Wellington, 69 Gracefield Road, Gracefield, Lower Hutt, 5010, New Zealand
| | - Vern L. Schramm
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| |
Collapse
|
26
|
The essential role of methylthioadenosine phosphorylase in prostate cancer. Oncotarget 2018; 7:14380-93. [PMID: 26910893 PMCID: PMC4924722 DOI: 10.18632/oncotarget.7486] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 01/24/2016] [Indexed: 11/25/2022] Open
Abstract
Prostatic epithelial cells secrete high levels of acetylated polyamines into the prostatic lumen. This distinctive characteristic places added strain on the connected pathways, which are forced to increase metabolite production to maintain pools. The methionine salvage pathway recycles the one-carbon unit lost to polyamine biosynthesis back to the methionine cycle, allowing for replenishment of SAM pools providing a mechanism to help mitigate metabolic stress associated with high flux through these pathways. The rate-limiting enzyme involved in this process is methylthioadenosine phosphorylase (MTAP), which, although commonly deleted in many cancers, is protected in prostate cancer. We report near universal retention of MTAP expression in a panel of human prostate cancer cell lines as well as patient samples. Upon metabolic perturbation, prostate cancer cell lines upregulate MTAP and this correlates with recovery of SAM levels. Furthermore, in a mouse model of prostate cancer we find that both normal prostate and diseased prostate maintain higher SAM levels than other tissues, even under increased metabolic stress. Finally, we show that knockdown of MTAP, both genetically and pharmacologically, blocks androgen sensitive prostate cancer growth in vivo. Our findings strongly suggest that the methionine salvage pathway is a major player in homeostatic regulation of metabolite pools in prostate cancer due to their high level of flux through the polyamine biosynthetic pathway. Therefore, this pathway, and specifically the MTAP enzyme, is an attractive therapeutic target for prostate cancer.
Collapse
|
27
|
Firestone RS, Schramm VL. The Transition-State Structure for Human MAT2A from Isotope Effects. J Am Chem Soc 2017; 139:13754-13760. [PMID: 28880543 DOI: 10.1021/jacs.7b05803] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Human methionine S-adenosyltransferase (MAT2A) catalyzes the formation of S-adenosylmethionine (SAM) from ATP and methionine. Synthetic lethal genetic analysis has identified MAT2A as an anticancer target in tumor cells lacking expression of 5'-methylthioadenosine phosphorylase (MTAP). Approximately 15% of human cancers are MTAP-/-. The remainder can be rendered MTAP- through MTAP inhibitors. We used kinetic isotope effect (KIE), commitment factor (Cf), and binding isotope effect (BIE) measurements combined with quantum mechanical (QM) calculations to solve the transition state structure of human MAT2A. The reaction is characterized by an advanced SN2 transition state. The bond forming from the nucleophilic methionine sulfur to the 5'-C of ATP is 2.03 Å at the transition state (bond order of 0.67). Departure of the leaving group triphosphate of ATP is well advanced and forms a 2.32 Å bond between the 5'-C of ATP and the oxygen of the triphosphate (bond order of 0.23). Interaction of MAT2A with its MAT2B regulatory subunit causes no change in the intrinsic KIEs, indicating the same transition state structure. The transition state for MAT2A is more advanced along the reaction coordinate (more product-like) than that from the near-symmetrical transition state of methionine adenosyltransferase from E. coli.
Collapse
Affiliation(s)
- Ross S Firestone
- Department of Biochemistry, Albert Einstein College of Medicine , 1300 Morris Park Avenue, Bronx, New York, New York 10461, United States
| | - Vern L Schramm
- Department of Biochemistry, Albert Einstein College of Medicine , 1300 Morris Park Avenue, Bronx, New York, New York 10461, United States
| |
Collapse
|
28
|
Bouton J, Van Hecke K, Van Calenbergh S. Efficient diastereoselective synthesis of a new class of azanucleosides: 2'-homoazanucleosides. Tetrahedron 2017; 73:4307-4316. [PMID: 32287431 PMCID: PMC7111761 DOI: 10.1016/j.tet.2017.05.083] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 05/24/2017] [Accepted: 05/27/2017] [Indexed: 02/08/2023]
Abstract
Azanucleosides, sugar-modified nucleoside analogues containing a 4' nitrogen atom, have shown a lot of therapeutic potential, e.g. as anti-cancer and antiviral agents. We report the synthesis of a series of 2'-homoazanucleosides, in which the nucleobase is attached to the 2'-position of the pyrrolidine ring via a methylene linker. A suitable orthogonally protected iminosugar was synthesized by ring closing metathesis and dihydroxylation as key steps and further converted to a series of 8 nucleoside analogues through Mitsunobu reaction with suitably protected nucleobases. The 5' position of the adenine analogue was then further derivatized with thiols to afford 2 additional compounds. The final compounds were evaluated for biological activity.
Collapse
Affiliation(s)
- Jakob Bouton
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Kristof Van Hecke
- XStruct, Department of Inorganic and Physical Chemistry, Ghent University, Krijgslaan 281 S3, 9000 Ghent, Belgium
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| |
Collapse
|
29
|
Jeong YJ, Cho HJ, Chung FL, Wang X, Hoe HS, Park KK, Kim CH, Chang HW, Lee SR, Chang YC. Isothiocyanates suppress the invasion and metastasis of tumors by targeting FAK/MMP-9 activity. Oncotarget 2017; 8:63949-63962. [PMID: 28969043 PMCID: PMC5609975 DOI: 10.18632/oncotarget.19213] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 06/10/2017] [Indexed: 01/03/2023] Open
Abstract
Isothiocyanates, which are present as glucosinolate precursors in cruciferous vegetables, have strong activity against various cancers. Here, we compared the anti-metastatic effects of isothiocyanates (benzyl isothiocyanate (BITC), phenethyl isothiocyanate (PEITC), and sulforaphane (SFN)) by examining how they regulate MMP-9 expression. Isothiocyanates, particularly PEITC, suppressed 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced MMP-9 activity and invasion in various cancer cell lines. By contrast, N-methyl phenethylamine, a PEITC analog without an isothiocyanate functional group, had no effect. A reporter gene assay demonstrated that BITC, PEITC, and SFN suppressed TAP-induced MMP-9 expression by inhibiting AP-1 and NF-κB in U20S osteosarcoma cells. All three compounds reduced phosphorylation of FAK, ERK1/2, and Akt. In addition, MMP-9 expression was downregulated by inhibiting FAK, ERK1/2, and Akt. Isothiocyanates-mediated inhibition of FAK phosphorylation suppressed phosphorylation of ERK1/2 and Akt in U2OS and A549 cells, along with the translocation of p65 and c-Fos, suggesting that isothiocyanates inhibit MMP-9 expression and cell invasion by blocking phosphorylation of FAK. Furthermore, isothiocyanates, abolished MMP-9 expression and tumor metastasis in vivo with the following efficacy: PEITC>BITC>SFN. Thus, isothiocyanates act as anti-metastatic compounds that suppress MMP-9 activity/expression by inhibiting NF-κB and AP-1 via suppression of the FAK/ERK and FAK/Akt signaling pathways.
Collapse
Affiliation(s)
- Yun-Jeong Jeong
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu 705-718, Republic of Korea
| | - Hyun-Ji Cho
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu 705-718, Republic of Korea.,Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu 701-300, Republic of Korea
| | - Fung-Lung Chung
- Department of Oncology, Lambardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Xiantao Wang
- Department of Oncology, Lambardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.,National Institutes of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hyang-Sook Hoe
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu 701-300, Republic of Korea
| | - Kwan-Kyu Park
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu 705-718, Republic of Korea
| | - Cheorl-Ho Kim
- Department of Biological Science, Sungkyunkwan University, Suwon, Kyunggi-Do 440-746, Republic of Korea
| | - Hyeun-Wook Chang
- College of pharmacy, Yeungnam University, Gyeongsan 701-947, Republic of Korea
| | - Sang-Rae Lee
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungbuk 28116, Republic of Korea
| | - Young-Chae Chang
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu 705-718, Republic of Korea
| |
Collapse
|
30
|
Firestone RS, Cameron SA, Karp JM, Arcus VL, Schramm VL. Heat Capacity Changes for Transition-State Analogue Binding and Catalysis with Human 5'-Methylthioadenosine Phosphorylase. ACS Chem Biol 2017; 12:464-473. [PMID: 28026167 PMCID: PMC5462123 DOI: 10.1021/acschembio.6b00885] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Human 5'-methylthioadenosine phosphorylase (MTAP) catalyzes the phosphorolysis of 5'-methylthioadenosine (MTA). Its action regulates cellular MTA and links polyamine synthesis to S-adenosylmethionine (AdoMet) salvage. Transition state analogues with picomolar dissociation constants bind to MTAP in an entropically driven process at physiological temperatures, suggesting increased hydrophobic character or dynamic structure for the complexes. Inhibitor binding exhibits a negative heat capacity change (-ΔCp), and thus the changes in enthalpy and entropy upon binding are strongly temperature-dependent. The ΔCp of inhibitor binding by isothermal titration calorimetry does not follow conventional trends and is contrary to that expected from the hydrophobic effect. Thus, ligands of increasing hydrophobicity bind with increasing values of ΔCp. Crystal structures of MTAP complexed to transition-state analogues MT-DADMe-ImmA, BT-DADMe-ImmA, PrT-ImmA, and a substrate analogue, MT-tubercidin, reveal similar active site contacts and overall protein structural parameters, despite large differences in ΔCp for binding. In addition, ΔCp values are not correlated with Kd values. Temperature dependence of presteady state kinetics revealed the chemical step for the MTAP reaction to have a negative heat capacity for transition state formation (-ΔCp‡). A comparison of the ΔCp‡ for MTAP presteady state chemistry and ΔCp for inhibitor binding revealed those transition-state analogues most structurally and thermodynamically similar to the transition state. Molecular dynamics simulations of MTAP apoenzyme and complexes with MT-DADMe-ImmA and MT-tubercidin show small, but increased dynamic motion in the inhibited complexes. Variable temperature CD spectroscopy studies for MTAP-inhibitor complexes indicate remarkable protein thermal stability (to Tm = 99 °C) in complexes with transition-state analogues.
Collapse
Affiliation(s)
- Ross S. Firestone
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Scott A. Cameron
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Jerome M. Karp
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Vickery L. Arcus
- Faculty of Science and Engineering, Department of Biological Science, University of Waikato, Private Bag 3105, Hamilton, New Zealand
| | - Vern L. Schramm
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States,Corresponding Author: Phone: 718-430-2813.
| |
Collapse
|
31
|
Firestone RS, Cameron SA, Tyler PC, Ducati RG, Spitz AZ, Schramm VL. Continuous Fluorescence Assays for Reactions Involving Adenine. Anal Chem 2016; 88:11860-11867. [PMID: 27779859 PMCID: PMC5434977 DOI: 10.1021/acs.analchem.6b03621] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
5'-Methylthioadenosine phosphorylase (MTAP) and 5'-methylthioadenosine nucleosidase (MTAN) catalyze the phosphorolysis and hydrolysis of 5'-methylthioadenosine (MTA), respectively. Both enzymes have low KM values for their substrates. Kinetic assays for these enzymes are challenging, as the ultraviolet absorbance spectra for reactant MTA and product adenine are similar. We report a new assay using 2-amino-5'-methylthioadenosine (2AMTA) as an alternative substrate for MTAP and MTAN enzymes. Hydrolysis or phosphorolysis of 2AMTA forms 2,6-diaminopurine, a fluorescent and easily quantitated product. We kinetically characterize 2AMTA with human MTAP, bacterial MTANs and use 2,6-diaminopurine as a fluorescent substrate for yeast adenine phosphoribosyltransferase. 2AMTA was used as the substrate to kinetically characterize the dissociation constants for three-transition-state analogue inhibitors of MTAP and MTAN. Kinetic values obtained from continuous fluorescent assays with MTA were in good agreement with previously measured literature values, but gave smaller experimental errors. Chemical synthesis from ribose and 2,6-dichloropurine provided crystalline 2AMTA as the oxalate salt. Chemo-enzymatic synthesis from ribose and 2,6-diaminopurine produced 2-amino-S-adenosylmethionine for hydrolytic conversion to 2AMTA. Interaction of 2AMTA with human MTAP was also characterized by pre-steady-state kinetics and by analysis of the crystal structure in a complex with sulfate as a catalytically inert analogue of phosphate. This assay is suitable for inhibitor screening by detection of fluorescent product, for quantitative analysis of hits by rapid and accurate measurement of inhibition constants in continuous assays, and pre-steady-state kinetic analysis of the target enzymes.
Collapse
Affiliation(s)
- Ross S. Firestone
- Department of Biochemistry, Albert Einstein College of
Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Scott A. Cameron
- Department of Biochemistry, Albert Einstein College of
Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Peter C. Tyler
- The Ferrier Research Institute, Victoria University of
Wellington, Lower Hutt, Wellington 6140, New Zealand
| | - Rodrigo G. Ducati
- Department of Biochemistry, Albert Einstein College of
Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Adam Z. Spitz
- Department of Biochemistry, Albert Einstein College of
Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Vern L. Schramm
- Department of Biochemistry, Albert Einstein College of
Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| |
Collapse
|
32
|
Marjon K, Cameron MJ, Quang P, Clasquin MF, Mandley E, Kunii K, McVay M, Choe S, Kernytsky A, Gross S, Konteatis Z, Murtie J, Blake ML, Travins J, Dorsch M, Biller SA, Marks KM. MTAP Deletions in Cancer Create Vulnerability to Targeting of the MAT2A/PRMT5/RIOK1 Axis. Cell Rep 2016; 15:574-587. [PMID: 27068473 DOI: 10.1016/j.celrep.2016.03.043] [Citation(s) in RCA: 277] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/08/2016] [Accepted: 03/10/2016] [Indexed: 12/31/2022] Open
Abstract
Homozygous deletions of p16/CDKN2A are prevalent in cancer, and these mutations commonly involve co-deletion of adjacent genes, including methylthioadenosine phosphorylase (MTAP). Here, we used shRNA screening and identified the metabolic enzyme, methionine adenosyltransferase II alpha (MAT2A), and the arginine methyltransferase, PRMT5, as vulnerable enzymes in cells with MTAP deletion. Metabolomic and biochemical studies revealed a mechanistic basis for this synthetic lethality. The MTAP substrate methylthioadenosine (MTA) accumulates upon MTAP loss. Biochemical profiling of a methyltransferase enzyme panel revealed that MTA is a potent and selective inhibitor of PRMT5. MTAP-deleted cells have reduced PRMT5 methylation activity and increased sensitivity to PRMT5 depletion. MAT2A produces the PRMT5 substrate S-adenosylmethionine (SAM), and MAT2A depletion reduces growth and PRMT5 methylation activity selectively in MTAP-deleted cells. Furthermore, this vulnerability extends to PRMT5 co-complex proteins such as RIOK1. Thus, the unique biochemical features of PRMT5 create an axis of targets vulnerable in CDKN2A/MTAP-deleted cancers.
Collapse
Affiliation(s)
- Katya Marjon
- Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA 02139, USA
| | | | - Phong Quang
- Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA 02139, USA
| | | | - Everton Mandley
- Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA 02139, USA
| | - Kaiko Kunii
- Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA 02139, USA
| | - Michael McVay
- Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA 02139, USA
| | - Sung Choe
- Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA 02139, USA
| | - Andrew Kernytsky
- Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA 02139, USA
| | - Stefan Gross
- Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA 02139, USA
| | - Zenon Konteatis
- Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA 02139, USA
| | - Joshua Murtie
- Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA 02139, USA
| | - Michelle L Blake
- Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA 02139, USA
| | - Jeremy Travins
- Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA 02139, USA
| | - Marion Dorsch
- Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA 02139, USA
| | - Scott A Biller
- Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA 02139, USA
| | - Kevin M Marks
- Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA 02139, USA.
| |
Collapse
|
33
|
Evans GB, Cameron SA, Luxenburger A, Guan R, Suarez J, Thomas K, Schramm VL, Tyler PC. Tight binding enantiomers of pre-clinical drug candidates. Bioorg Med Chem 2015; 23:5326-33. [PMID: 26260335 PMCID: PMC4567382 DOI: 10.1016/j.bmc.2015.07.059] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 07/26/2015] [Accepted: 07/27/2015] [Indexed: 11/30/2022]
Abstract
MTDIA is a picomolar transition state analogue inhibitor of human methylthioadenosine phosphorylase and a femtomolar inhibitor of Escherichia coli methylthioadenosine nucleosidase. MTDIA has proven to be a non-toxic, orally available pre-clinical drug candidate with remarkable anti-tumour activity against a variety of human cancers in mouse xenografts. The structurally similar compound MTDIH is a potent inhibitor of human and malarial purine nucleoside phosphorylase (PNP) as well as the newly discovered enzyme, methylthioinosine phosphorylase, isolated from Pseudomonas aeruginosa. Since the enantiomers of some pharmaceuticals have revealed surprising biological activities, the enantiomers of MTDIH and MTDIA, compounds 1 and 2, respectively, were prepared and their enzyme binding properties studied. Despite binding less tightly to their target enzymes than their enantiomers compounds 1 and 2 are nanomolar inhibitors.
Collapse
Affiliation(s)
- Gary B Evans
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Rd, Lower Hutt 5010, New Zealand
| | - Scott A Cameron
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Rd, Lower Hutt 5010, New Zealand; Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA
| | - Andreas Luxenburger
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Rd, Lower Hutt 5010, New Zealand
| | - Rong Guan
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA
| | - Javier Suarez
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA
| | - Keisha Thomas
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA
| | - Vern L Schramm
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA
| | - Peter C Tyler
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Rd, Lower Hutt 5010, New Zealand
| |
Collapse
|
34
|
Li CF, Fang FM, Kung HJ, Chen LT, Wang JW, Tsai JW, Yu SC, Wang YH, Li SH, Huang HY. Downregulated MTAP expression in myxofibrosarcoma: A characterization of inactivating mechanisms, tumor suppressive function, and therapeutic relevance. Oncotarget 2015; 5:11428-41. [PMID: 25426549 PMCID: PMC4294342 DOI: 10.18632/oncotarget.2552] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 10/01/2014] [Indexed: 12/22/2022] Open
Abstract
Myxofibrosarcomas are genetically complex and involve recurrently deleted chromosome 9p, for which we characterized the pathogenically relevant target(s) using genomic profiling. In 12 of the 15 samples, we detected complete or partial losses of 9p. The only aggressiveness-associated, differentially lost region was 9p21.3, spanning the potential inactivated methylthioadenosine phosphorylase (MTAP) that exhibited homozygous (4/15) or hemizygous (3/15) deletions. In independent samples, MTAP gene status was assessed using quantitative- and methylation-specific PCR assays, and immunoexpression was evaluated. We applied MTAP reexpression or knockdown to elucidate the functional roles of MTAP and the therapeutic potential of L-alanosine in MTAP-preserved and MTAP-deficient myxofibrosarcoma cell lines and xenografts. MTAP protein deficiency (37%) was associated with MTAP gene inactivation (P < 0.001) by homozygous deletion or promoter methylation, and independently portended unfavorable metastasis-free survival (P = 0.0318) and disease-specific survival (P = 0.014). Among the MTAP-deficient cases, the homozygous deletion of MTAP predicted adverse outcome. In MTAP-deficient cells, MTAP reexpression inhibited cell migration and invasion, proliferation, and anchorage-independent colony formation and downregulated cyclin D1. This approach also attenuated the tube-forming abilities of human umbilical venous endothelial cells, attributable to the transcriptional repression of MMP-9, and abrogated the susceptibility to L-alanosine. The inhibiting effects of MTAP expression on tumor growth, angiogenesis, and the induction of apoptosis by L-alanosine were validated using MTAP-reexpressing xenografts and reverted using RNA interference in MTAP-preserved cells. In conclusion, homozygous deletion primarily accounts for the adverse prognostic impact of MTAP deficiency and confers the biological aggressiveness and susceptibility to L-alanosine in myxofibrosarcomas.
Collapse
Affiliation(s)
- Chien-Feng Li
- Department of Pathology, Chi-Mei Medical Center, Tainan, Taiwan. Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan. National Institute of Cancer Research National Health Research Institutes, Tainan, Taiwan. Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Fu-Min Fang
- Departments of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hsing-Jien Kung
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Tainan, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research National Health Research Institutes, Tainan, Taiwan. Department of Internal Medicine and Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jun-Wen Wang
- Orthopedic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Jen-Wei Tsai
- Department of Anatomic Pathology, E-Da Hospital, Kaohsiung, Tawian
| | - Shih Chen Yu
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Hui Wang
- Institute of Biosignal Transduction, National Cheng Kung University, Tainan, Taiwan
| | - Shau-Hsuan Li
- Department of Internal Medicine, Division of Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hsuan-Ying Huang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
35
|
Lee CJ, Lee MH, Yoo SM, Choi KI, Song JH, Jang JH, Oh SR, Ryu HW, Lee HS, Surh YJ, Cho YY. Magnolin inhibits cell migration and invasion by targeting the ERKs/RSK2 signaling pathway. BMC Cancer 2015; 15:576. [PMID: 26253302 PMCID: PMC4529708 DOI: 10.1186/s12885-015-1580-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 07/27/2015] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Magnolin is a natural compound abundantly found in Magnolia flos, which has been traditionally used in oriental medicine to treat headaches, nasal congestion and anti-inflammatory reactions. Our recent results have demonstrated that magnolin targets the active pockets of ERK1 and ERK2, which are important signaling molecules in cancer cell metastasis. The aim of this study is to evaluate the effects of magnolin on cell migration and to further explore the molecular mechanisms involved. METHODS Magnolin-mediated signaling inhibition was confirmed by Western blotting using RSK2(+/+) and RSK2(-/-) MEFs, A549 and NCI-H1975 lung cancer cells, and by NF-κB and Cox-2 promoter luciferase reporter assays. Inhibition of cell migration by magnolin was examined by wound healing and/or Boyden Chamber assays using JB6 Cl41 and A549 human lung cancer cells. The molecular mechanisms involved in cell migration and epithelial-to-mesenchymal transition were determined by zymography, Western blotting, real-time PCR and immunocytofluorescence. RESULTS Magnolin inhibited NF-κB transactivation activity by suppressing the ERKs/RSK2 signaling pathway. Moreover, magnolin abrogated the increase in EGF-induced COX-2 protein levels and wound healing. In human lung cancer cells such as A549 and NCI-H1975, which harbor constitutive active Ras and EGFR mutants, respectively, magnolin suppressed wound healing and cell invasion as seen by a Boyden chamber assay. In addition, it was observed that magnolin inhibited MMP-2 and -9 gene expression and activity. The knockdown or knockout of RSK2 in A549 lung cancer cells or MEFs revealed that magnolin targeting ERKs/RSK2 signaling suppressed epithelial-to-mesenchymal transition by modulating EMT marker proteins such as N-cadherin, E-cadherin, Snail, Vimentin and MMPs. CONCLUSIONS These results demonstrate that magnolin inhibits cell migration and invasion by targeting the ERKs/RSK2 signaling pathway.
Collapse
Affiliation(s)
- Cheol-Jung Lee
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 420-743, Republic of Korea.
| | - Mee-Hyun Lee
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 420-743, Republic of Korea.
| | - Sun-Mi Yoo
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 420-743, Republic of Korea.
| | - Kyung-Il Choi
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 420-743, Republic of Korea.
| | - Ji-Hong Song
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 420-743, Republic of Korea.
| | - Jeong-Hoon Jang
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 420-743, Republic of Korea. .,College of Pharmacy, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 151-742, Republic of Korea.
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gun, ChungBuk, 363-883, Republic of Korea.
| | - Hyung-Won Ryu
- Natural Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gun, ChungBuk, 363-883, Republic of Korea.
| | - Hye-Suk Lee
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 420-743, Republic of Korea.
| | - Young-Joon Surh
- College of Pharmacy, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 151-742, Republic of Korea.
| | - Yong-Yeon Cho
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 420-743, Republic of Korea.
| |
Collapse
|
36
|
Sprenger J, Svensson B, Hålander J, Carey J, Persson L, Al-Karadaghi S. Three-dimensional structures of Plasmodium falciparum spermidine synthase with bound inhibitors suggest new strategies for drug design. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2015; 71:484-93. [PMID: 25760598 PMCID: PMC4356361 DOI: 10.1107/s1399004714027011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 12/09/2014] [Indexed: 01/06/2023]
Abstract
The enzymes of the polyamine-biosynthesis pathway have been proposed to be promising drug targets in the treatment of malaria. Spermidine synthase (SpdS; putrescine aminopropyltransferase) catalyzes the transfer of the aminopropyl moiety from decarboxylated S-adenosylmethionine to putrescine, leading to the formation of spermidine and 5'-methylthioadenosine (MTA). In this work, X-ray crystallography was used to examine ligand complexes of SpdS from the malaria parasite Plasmodium falciparum (PfSpdS). Five crystal structures were determined of PfSpdS in complex with MTA and the substrate putrescine, with MTA and spermidine, which was obtained as a result of the enzymatic reaction taking place within the crystals, with dcAdoMet and the inhibitor 4-methylaniline, with MTA and 4-aminomethylaniline, and with a compound predicted in earlier in silico screening to bind to the active site of the enzyme, benzimidazol-(2-yl)pentan-1-amine (BIPA). In contrast to the other inhibitors tested, the complex with BIPA was obtained without any ligand bound to the dcAdoMet-binding site of the enzyme. The complexes with the aniline compounds and BIPA revealed a new mode of ligand binding to PfSpdS. The observed binding mode of the ligands, and the interplay between the two substrate-binding sites and the flexible gatekeeper loop, can be used in the design of new approaches in the search for new inhibitors of SpdS.
Collapse
Affiliation(s)
- Janina Sprenger
- Center for Molecular Protein Science, Lund University, SE-221 00 Lund, Sweden
- Department of Experimental Medical Science, Lund University, SE-221 84 Lund, Sweden
| | - Bo Svensson
- Center for Molecular Protein Science, Lund University, SE-221 00 Lund, Sweden
- SARomics Biostructures AB, Box 724, SE-220 07 Lund, Sweden
| | - Jenny Hålander
- Center for Molecular Protein Science, Lund University, SE-221 00 Lund, Sweden
| | - Jannette Carey
- Chemistry Department, Princeton University, Princeton, New Jersey, USA
| | - Lo Persson
- Department of Experimental Medical Science, Lund University, SE-221 84 Lund, Sweden
| | - Salam Al-Karadaghi
- Center for Molecular Protein Science, Lund University, SE-221 00 Lund, Sweden
| |
Collapse
|
37
|
Wikoff WR, Grapov D, Fahrmann JF, DeFelice B, Rom WN, Pass HI, Kim K, Nguyen U, Taylor SL, Gandara DR, Kelly K, Fiehn O, Miyamoto S. Metabolomic markers of altered nucleotide metabolism in early stage adenocarcinoma. Cancer Prev Res (Phila) 2015; 8:410-8. [PMID: 25657018 DOI: 10.1158/1940-6207.capr-14-0329] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/29/2015] [Indexed: 12/25/2022]
Abstract
Adenocarcinoma, a type of non-small cell lung cancer, is the most frequently diagnosed lung cancer and the leading cause of lung cancer mortality in the United States. It is well documented that biochemical changes occur early in the transition from normal to cancer cells, but the extent to which these alterations affect tumorigenesis in adenocarcinoma remains largely unknown. Herein, we describe the application of mass spectrometry and multivariate statistical analysis in one of the largest biomarker research studies to date aimed at distinguishing metabolic differences between malignant and nonmalignant lung tissue. Gas chromatography time-of-flight mass spectrometry was used to measure 462 metabolites in 39 malignant and nonmalignant lung tissue pairs from current or former smokers with early stage (stage IA-IB) adenocarcinoma. Statistical mixed effects models, orthogonal partial least squares discriminant analysis and network integration, were used to identify key cancer-associated metabolic perturbations in adenocarcinoma compared with nonmalignant tissue. Cancer-associated biochemical alterations were characterized by (i) decreased glucose levels, consistent with the Warburg effect, (ii) changes in cellular redox status highlighted by elevations in cysteine and antioxidants, alpha- and gamma-tocopherol, (iii) elevations in nucleotide metabolites 5,6-dihydrouracil and xanthine suggestive of increased dihydropyrimidine dehydrogenase and xanthine oxidoreductase activity, (iv) increased 5'-deoxy-5'-methylthioadenosine levels indicative of reduced purine salvage and increased de novo purine synthesis, and (v) coordinated elevations in glutamate and UDP-N-acetylglucosamine suggesting increased protein glycosylation. The present study revealed distinct metabolic perturbations associated with early stage lung adenocarcinoma, which may provide candidate molecular targets for personalizing therapeutic interventions and treatment efficacy monitoring.
Collapse
Affiliation(s)
- William R Wikoff
- University of California, Davis Genome Center, Davis, California
| | - Dmitry Grapov
- University of California, Davis Genome Center, Davis, California
| | | | - Brian DeFelice
- University of California, Davis Genome Center, Davis, California
| | - William N Rom
- Division of Pulmonary, Critical Care and Sleep, New York University, School of Medicine New York, New York
| | - Harvey I Pass
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Langone Medical Center, New York University, New York, New York
| | - Kyoungmi Kim
- Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California Davis, Davis, California
| | - UyenThao Nguyen
- Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California Davis, Davis, California
| | - Sandra L Taylor
- Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California Davis, Davis, California
| | - David R Gandara
- University of California, Davis Genome Center, Davis, California. Division of Pulmonary, Critical Care and Sleep, New York University, School of Medicine New York, New York. Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Langone Medical Center, New York University, New York, New York. Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California Davis, Davis, California. Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California, Davis Medical Center, Sacramento, California. Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi-Arabia
| | - Karen Kelly
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California, Davis Medical Center, Sacramento, California
| | - Oliver Fiehn
- University of California, Davis Genome Center, Davis, California. Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi-Arabia
| | - Suzanne Miyamoto
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California, Davis Medical Center, Sacramento, California.
| |
Collapse
|
38
|
Brown-Borg HM, Rakoczy S, Wonderlich JA, Armstrong V, Rojanathammanee L. Altered dietary methionine differentially impacts glutathione and methionine metabolism in long-living growth hormone-deficient Ames dwarf and wild-type mice. LONGEVITY & HEALTHSPAN 2014; 3:10. [PMID: 25584190 PMCID: PMC4290132 DOI: 10.1186/2046-2395-3-10] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/01/2014] [Indexed: 02/05/2023]
Abstract
Background Extending mammalian health span and life span has been achieved under a variety of dietary restriction protocols. Reducing the intake of a specific amino acid has also been shown to extend health and longevity. We recently reported that methionine (MET) restriction is not effective in life span extension in growth hormone (GH) signaling mutants. To better understand the apparent necessity of GH in the ‘sensing’ of altered dietary MET, the current study was designed to evaluate MET and glutathione (GSH) metabolism (as well as other pathways) in long-living GH-deficient Ames dwarf and wild-type mice following 8 weeks of restricted (0.16%), low (0.43%), or enriched (1.3%) dietary MET consumption. Metabolite expression was examined in liver tissue, while gene and protein expression were evaluated in liver, kidney, and muscle tissues. Results Body weight was maintained in dwarf mice on the MET diets, while wild-type mice on higher levels of MET gained weight. Liver MET levels were similar in Ames mice, while several MET pathway enzymes were elevated regardless of dietary MET intake. Transsulfuration enzymes were also elevated in Ames mice but differences in cysteine levels were not different between genotypes. Dwarf mice maintained higher levels of GSH on MET restriction compared to wild-type mice, while genotype and diet effects were also detected in thioredoxin and glutaredoxin. MET restriction increased transmethylation in both genotypes as indicated by increased S-adenosylmethionine (SAM), betaine, and dimethylglycine. Diet did not impact levels of glycolytic components, but dwarf mice exhibited higher levels of key members of this pathway. Coenzyme A and measures of fatty acid oxidation were elevated in dwarf mice and unaffected by diet. Conclusions This component analysis between Ames and wild-type mice suggests that the life span differences observed may result from the atypical MET metabolism and downstream effects on multiple systems. The overall lack of responsiveness to the different diets is well reflected across many metabolic pathways in dwarf mice indicating the importance of GH signaling in the ability to discriminate dietary amino acid levels. Electronic supplementary material The online version of this article (doi:10.1186/2046-2395-3-10) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Holly M Brown-Borg
- Department of Basic Sciences, University of North Dakota School of Medicine & Health Sciences, 501 N. Columbia Road, Grand Forks, ND 58203 USA
| | - Sharlene Rakoczy
- Department of Basic Sciences, University of North Dakota School of Medicine & Health Sciences, 501 N. Columbia Road, Grand Forks, ND 58203 USA
| | - Joseph A Wonderlich
- Department of Basic Sciences, University of North Dakota School of Medicine & Health Sciences, 501 N. Columbia Road, Grand Forks, ND 58203 USA
| | - Vanessa Armstrong
- Department of Basic Sciences, University of North Dakota School of Medicine & Health Sciences, 501 N. Columbia Road, Grand Forks, ND 58203 USA
| | - Lalida Rojanathammanee
- Department of Basic Sciences, University of North Dakota School of Medicine & Health Sciences, 501 N. Columbia Road, Grand Forks, ND 58203 USA ; School of Sports Science, Institute of Science, Suranaree University of Technology, Muang District, Nakhon Ratchasima, 30000 Thailand
| |
Collapse
|
39
|
MTAP is an independent prognosis marker and the concordant loss of MTAP and p16 expression predicts short survival in non-small cell lung cancer patients. Eur J Surg Oncol 2014; 40:1143-50. [PMID: 24969958 DOI: 10.1016/j.ejso.2014.04.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/24/2014] [Accepted: 04/24/2014] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Methylthioadenosine phosphorylase (MTAP), a ubiquitously expressed protein, plays important roles in purine biosynthesis. Locating near to each other on chromosome 9p21-22, codeletion of the MTAP and p16(Ink4A) genes have been reported in non-small cell lung cancer (NSCLC). The aim of this study is to determine the respective prognostic value of MTAP and p16 by considering their correlation in NSCLC patients. MATERIALS AND METHODS We analyzed MTAP and p16 protein expression by immunohistochemical staining on 99 NSCLC tissue microarray samples. The association between MTAP and p16 expression levels and prognosis were analyzed using the Kaplan-Meier method and Cox proportional hazards model for prognosis. RESULTS Patients with a low MTAP expression level had poor overall survival (P = 0.010) and disease-free survival (P = 0.002). Low p16 expression indicated a trend toward poor overall survival (P = 0.138) and disease-free survival (P = 0.199). There was a significant positive correlation between MTAP and p16 expression levels (Spearman's ρ = 0.402, P < 0.001). By multivariate analyses, the MTAP expression level retained its independent prognostic power and p16 expression loss of the correlation with prognosis. Concordant loss of MTAP and p16 expression was observed in 24 out of 99 patients (24.2%). Patients with concordant loss of MTAP and p16 expression had the worst prognosis compared to patients with high expression of both markers. CONCLUSION MTAP expression is an independent prognostic factor and has greater prognostic significance than p16 expression in NSCLC. Concordant loss of MTAP and p16 expression indicates poor outcomes in lung cancer patients.
Collapse
|
40
|
Shlomi T, Fan J, Tang B, Kruger WD, Rabinowitz JD. Quantitation of cellular metabolic fluxes of methionine. Anal Chem 2014; 86:1583-91. [PMID: 24397525 DOI: 10.1021/ac4032093] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Methionine is an essential proteogenic amino acid. In addition, it is a methyl donor for DNA and protein methylation and a propylamine donor for polyamine biosynthesis. Both the methyl and propylamine donation pathways involve metabolic cycles, and methods are needed to quantitate these cycles. Here, we describe an analytical approach for quantifying methionine metabolic fluxes that accounts for the mixing of intracellular and extracellular methionine pools. We observe that such mixing prevents isotope tracing experiments from reaching the steady state due to the large size of the media pools and hence precludes the use of standard stationary metabolic flux analysis. Our approach is based on feeding cells with (13)C methionine and measuring the isotope-labeling kinetics of both intracellular and extracellular methionine by liquid chromatography-mass spectrometry (LC-MS). We apply this method to quantify methionine metabolism in a human fibrosarcoma cell line and study how methionine salvage pathway enzyme methylthioadenosine phosphorylase (MTAP), frequently deleted in cancer, affects methionine metabolism. We find that both transmethylation and propylamine transfer fluxes amount to roughly 15% of the net methionine uptake, with no major changes due to MTAP deletion. Our method further enables the quantification of flux through the pro-tumorigenic enzyme ornithine decarboxylase, and this flux increases 2-fold following MTAP deletion. The analytical approach used to quantify methionine metabolic fluxes is applicable for other metabolic systems affected by mixing of intracellular and extracellular metabolite pools.
Collapse
Affiliation(s)
- Tomer Shlomi
- Dept. of Computer Science, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | | | | | | | | |
Collapse
|
41
|
Guan R, Tyler PC, Evans GB, Schramm VL. Thermodynamic analysis of transition-state features in picomolar inhibitors of human 5'-methylthioadenosine phosphorylase. Biochemistry 2013; 52:8313-22. [PMID: 24148083 DOI: 10.1021/bi401188w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Human 5'-methylthioadenosine phosphorylase (MTAP) is solely responsible for 5'-methylthioadenosine (MTA) metabolism to permit S-adenosylmethionine salvage. Transition-state (TS) analogues of MTAP are in development as anticancer candidates. TS analogues of MTAP incorporate a cationic nitrogen and a protonated 9-deazaadenine leaving group, which are mimics of the ribocation transition state. MT-ImmA and MT-DADMe-ImmA are two examples of these TS analogues. Thermodynamic analysis of MTA, inhibitor, and phosphate binding reveals the cationic nitrogen to provide -2.6 and -3.6 kcal/mol binding free energy for MT-ImmA and MT-DADMe-ImmA, respectively. The protonated deazaadenine provides an additional -1.3 (MT-ImmA) to -1.7 kcal/mol (MT-DADMe-ImmA). MT-DADMe-ImmA is a better match in TS geometry than MT-ImmA and is thermodynamically favored. Binding of TS analogues to the MTAP/phosphate complex is fully entropic, in contrast to TS analogue binding to the related human purine nucleoside phosphorylase/phosphate complex, which is fully enthalpic (Guan, R., Ho, M. C., Brenowitz, M., Tyler, P. C., Evans, G. B., Almo, S. C., and Schramm, V. L. (2011) Biochemistry 50, 10408-10417). The binding thermodynamics of phosphate or TS analogues alone to MTAP are fully dominated by enthalpy. Phosphate anchored in the catalytic site forms an ion pair with the cationic TS analogue to cause stabilization of the enzyme structure in the ternary complex. The ternary-induced conformational changes convert the individual enthalpic binding energies to entropy, resulting in a presumed shift of the protein architecture toward the transition state. Formation of the ternary TS analogue complex with MTAP induces a remarkable increase in thermal stability (ΔTm 28 °C). The enthalpic, entropic, and protein-stability features of TS analogue binding to human MTAP are resolved in these studies.
Collapse
Affiliation(s)
- Rong Guan
- From the Department of Biochemistry, Albert Einstein College of Medicine, Yeshiva University , 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | | | | | | |
Collapse
|
42
|
Abstract
Enzymes achieve their transition states by dynamic conformational searches on the femtosecond to picosecond time scale. Mimics of reactants at enzymatic transition states bind tightly to enzymes by stabilizing the conformation optimized through evolution for transition state formation. Instead of forming the transient transition state geometry, transition state analogues convert the short-lived transition state to a stable thermodynamic state. Enzymatic transition states are understood by combining kinetic isotope effects and computational chemistry. Analogues of the transition state can bind millions of times more tightly than substrates and show promise for drug development for several targets.
Collapse
Affiliation(s)
- Vern L Schramm
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx New York 10461, United States.
| |
Collapse
|
43
|
Abstract
S-adenosylmethionine (AdoMet, also known as SAM and SAMe) is the principal biological methyl donor synthesized in all mammalian cells but most abundantly in the liver. Biosynthesis of AdoMet requires the enzyme methionine adenosyltransferase (MAT). In mammals, two genes, MAT1A that is largely expressed by normal liver and MAT2A that is expressed by all extrahepatic tissues, encode MAT. Patients with chronic liver disease have reduced MAT activity and AdoMet levels. Mice lacking Mat1a have reduced hepatic AdoMet levels and develop oxidative stress, steatohepatitis, and hepatocellular carcinoma (HCC). In these mice, several signaling pathways are abnormal that can contribute to HCC formation. However, injury and HCC also occur if hepatic AdoMet level is excessive chronically. This can result from inactive mutation of the enzyme glycine N-methyltransferase (GNMT). Children with GNMT mutation have elevated liver transaminases, and Gnmt knockout mice develop liver injury, fibrosis, and HCC. Thus a normal hepatic AdoMet level is necessary to maintain liver health and prevent injury and HCC. AdoMet is effective in cholestasis of pregnancy, and its role in other human liver diseases remains to be better defined. In experimental models, it is effective as a chemopreventive agent in HCC and perhaps other forms of cancer as well.
Collapse
Affiliation(s)
- Shelly C Lu
- Division of Gastroenterology and Liver Diseases, USC Research Center for Liver Diseases, Southern California Research Center for ALPD and Cirrhosis, Keck School of Medicine, Los Angeles, California 90033, USA.
| | | |
Collapse
|
44
|
Kim JB, Deluna A, Mungrue IN, Vu C, Pouldar D, Civelek M, Orozco L, Wu J, Wang X, Charugundla S, Castellani LW, Rusek M, Jakubowski H, Jakobowski H, Lusis AJ. Effect of 9p21.3 coronary artery disease locus neighboring genes on atherosclerosis in mice. Circulation 2012; 126:1896-906. [PMID: 22952318 DOI: 10.1161/circulationaha.111.064881] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND The human 9p21.3 chromosome locus has been shown to be an independent risk factor for atherosclerosis in multiple large-scale genome-wide association studies, but the underlying mechanism remains unknown. We set out to investigate the potential role of the 9p21.3 locus neighboring genes, including Mtap, the 2 isoforms of Cdkn2a, p16Ink4a and p19Arf, and Cdkn2b, in atherosclerosis using knockout mice models. METHODS AND RESULTS Gene-targeted mice for neighboring genes, including Mtap, Cdkn2a, p19Arf, and Cdkn2b, were each bred to mice carrying the human APO*E3 Leiden transgene that sensitizes the mice for atherosclerotic lesions through elevated plasma cholesterol. We found that the mice heterozygous for Mtap developed larger lesions compared with wild-type mice (49623±21650 versus 18899±9604 μm(2) per section [mean±SD]; P=0.01), with morphology similar to that of wild-type mice. The Mtap heterozygous mice demonstrated changes in metabolic and methylation profiles and CD4(+) cell counts. The Cdkn2a knockout mice had smaller lesions compared with wild-type and heterozygous mice, and there were no significant differences in lesion size in p19Arf and Cdkn2b mutants compared with wild type. We observed extensive, tissue-specific compensatory regulation of the Cdkn2a and Cdkn2b genes among the various knockout mice, making the effects on atherosclerosis difficult to interpret. CONCLUSIONS Mtap plays a protective role against atherosclerosis, whereas Cdkn2a appears to be modestly proatherogenic. However, no relation was found between the 9p21 genotype and the transcription of 9p21 neighboring genes in primary human aortic vascular cells in vitro. There is extensive compensatory regulation in the highly conserved 9p21 orthologous region in mice.
Collapse
Affiliation(s)
- Juyong Brian Kim
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1679, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Wang S, Haapalainen AM, Yan F, Du Q, Tyler PC, Evans GB, Rinaldo-Matthis A, Brown RL, Norris GE, Almo SC, Schramm VL. A picomolar transition state analogue inhibitor of MTAN as a specific antibiotic for Helicobacter pylori. Biochemistry 2012; 51:6892-4. [PMID: 22891633 DOI: 10.1021/bi3009664] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Campylobacter and Helicobacter species express a 6-amino-6-deoxyfutalosine N-ribosylhydrolase (HpMTAN) proposed to function in menaquinone synthesis. BuT-DADMe-ImmA is a 36 pM transition state analogue of HpMTAN, and the crystal structure of the enzyme-inhibitor complex reveals the mechanism of inhibition. BuT-DADMe-ImmA has a MIC(90) value of <8 ng/mL for Helicobacter pylori growth but does not cause growth arrest in other common clinical pathogens, thus demonstrating potential as an H. pylori-specific antibiotic.
Collapse
Affiliation(s)
- Shanzhi Wang
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Clinch K, Evans GB, Fröhlich RFG, Gulab SA, Gutierrez JA, Mason JM, Schramm VL, Tyler PC, Woolhouse AD. Transition state analogue inhibitors of human methylthioadenosine phosphorylase and bacterial methylthioadenosine/S-adenosylhomocysteine nucleosidase incorporating acyclic ribooxacarbenium ion mimics. Bioorg Med Chem 2012; 20:5181-7. [PMID: 22854195 DOI: 10.1016/j.bmc.2012.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 06/28/2012] [Accepted: 07/04/2012] [Indexed: 11/24/2022]
Abstract
Several acyclic hydroxy-methylthio-amines with 3-5 carbon atoms were prepared and coupled via a methylene link to 9-deazaadenine. The products were tested for inhibition against human MTAP and Escherichia coli and Neisseria meningitidis MTANs and gave K(i) values as low as 0.23 nM. These results were compared to those obtained with 1st and 2nd generation inhibitors (1S)-1-(9-deazaadenin-9-yl)-1,4-dideoxy-1,4-imino-5-methylthio-D-ribitol (MT-Immucillin-A, 3) and (3R,4S)-1-[9-deazaadenin-9-yl)methyl]3-hydroxy-4-methylthiomethylpyrrolidine (MT-DADMe-Immucillin-A, 4). The best inhibitors were found to exhibit binding affinities of approximately 2- to 4-fold those of 3 but were significantly weaker than 4. Cleavage of the 2,3 carbon-carbon bond in MT-Immucillin-A (3) gave an acyclic product (79) with a 21,500 fold loss of activity against E. coli MTAN. In another case, N-methylation of a side chain secondary amine resulted in a 250-fold loss of activity against the same enzyme [(±)-65 vs (±)-68]. The inhibition results were also contrasted with those acyclic derivatives previously prepared as inhibitors for a related enzyme, purine nucleoside phosphorylase (PNP), where some inhibitors in the latter case were found to be more potent than their cyclic counterparts.
Collapse
Affiliation(s)
- Keith Clinch
- Carbohydrate Chemistry, Industrial Research Limited, PO Box 31310, Lower Hutt 5040, New Zealand.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Cavuoto P, Fenech MF. A review of methionine dependency and the role of methionine restriction in cancer growth control and life-span extension. Cancer Treat Rev 2012; 38:726-36. [PMID: 22342103 DOI: 10.1016/j.ctrv.2012.01.004] [Citation(s) in RCA: 214] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 12/22/2011] [Accepted: 01/15/2012] [Indexed: 01/11/2023]
Abstract
Methionine is an essential amino acid with many key roles in mammalian metabolism such as protein synthesis, methylation of DNA and polyamine synthesis. Restriction of methionine may be an important strategy in cancer growth control particularly in cancers that exhibit dependence on methionine for survival and proliferation. Methionine dependence in cancer may be due to one or a combination of deletions, polymorphisms or alterations in expression of genes in the methionine de novo and salvage pathways. Cancer cells with these defects are unable to regenerate methionine via these pathways. Defects in the metabolism of folate may also contribute to the methionine dependence phenotype in cancer. Selective killing of methionine dependent cancer cells in co-culture with normal cells has been demonstrated using culture media deficient in methionine. Several animal studies utilizing a methionine restricted diet have reported inhibition of cancer growth and extension of a healthy life-span. In humans, vegan diets, which can be low in methionine, may prove to be a useful nutritional strategy in cancer growth control. The development of methioninase which depletes circulating levels of methionine may be another useful strategy in limiting cancer growth. The application of nutritional methionine restriction and methioninase in combination with chemotherapeutic regimens is the current focus of clinical studies.
Collapse
Affiliation(s)
- Paul Cavuoto
- CSIRO Food and Nutritional Sciences, P.O. Box 10041, Adelaide BC, SA 5000, Australia.
| | | |
Collapse
|
48
|
Hishiki T, Yamamoto T, Morikawa T, Kubo A, Kajimura M, Suematsu M. Carbon monoxide: impact on remethylation/transsulfuration metabolism and its pathophysiologic implications. J Mol Med (Berl) 2012; 90:245-54. [PMID: 22331189 PMCID: PMC3296020 DOI: 10.1007/s00109-012-0875-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 01/27/2012] [Accepted: 01/31/2012] [Indexed: 01/08/2023]
Abstract
Carbon monoxide (CO) is a gaseous product generated by heme oxygenase (HO), which oxidatively degrades heme. While the stress-inducible HO-1 has well been recognized as an anti-oxidative defense mechanism under stress conditions, recent studies suggest that cancer cells utilize the reaction for their survival. HO-2, the constitutive isozyme, also plays protective roles as a tonic regulator for neurovascular function. Although protective roles of the enzyme reaction and CO have extensively been studied, little information is available on the molecular mechanisms by which the gas exerts its biological actions. Recent studies using metabolomics revealed that CO inhibits cystathionine β-synthase (CBS), which generates H2S, another gaseous mediator. The CO-dependent CBS inhibition may impact on the remethylation cycle and related metabolic pathways including the methionine salvage pathway and polyamine synthesis. This review focuses on the gas-responsive regulation of metabolic systems, particularly the remethylation and transsulfuration pathways, and their putative implications for cancer and ischemic diseases.
Collapse
Affiliation(s)
- Takako Hishiki
- Department of Biochemistry, JST, ERATO, Suematsu Gas Biology Project, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | |
Collapse
|
49
|
Collins CC, Volik SV, Lapuk AV, Wang Y, Gout PW, Wu C, Xue H, Cheng H, Haegert A, Bell RH, Brahmbhatt S, Anderson S, Fazli L, Hurtado-Coll A, Rubin MA, Demichelis F, Beltran H, Hirst M, Marra M, Maher CA, Chinnaiyan AM, Gleave M, Bertino JR, Lubin M, Wang Y. Next generation sequencing of prostate cancer from a patient identifies a deficiency of methylthioadenosine phosphorylase, an exploitable tumor target. Mol Cancer Ther 2012; 11:775-83. [PMID: 22252602 DOI: 10.1158/1535-7163.mct-11-0826] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Castrate-resistant prostate cancer (CRPC) and neuroendocrine carcinoma of the prostate are invariably fatal diseases for which only palliative therapies exist. As part of a prostate tumor sequencing program, a patient tumor was analyzed using Illumina genome sequencing and a matched renal capsule tumor xenograft was generated. Both tumor and xenograft had a homozygous 9p21 deletion spanning the MTAP, CDKN2, and ARF genes. It is rare for this deletion to occur in primary prostate tumors, yet approximately 10% express decreased levels of methylthioadenosine phosphorylase (MTAP) mRNA. Decreased MTAP expression is a prognosticator for poor outcome. Moreover, it seems that this deletion is more common in CRPC than in primary prostate cancer. We show for the first time that treatment with methylthioadenosine and high dose 6-thioguanine causes marked inhibition of a patient-derived neuroendocrine xenograft growth while protecting the host from 6-thioguanine toxicity. This therapeutic approach can be applied to other MTAP-deficient human cancers as deletion or hypermethylation of the MTAP gene occurs in a broad spectrum of tumors at high frequency. The combination of genome sequencing and patient-derived xenografts can identify candidate therapeutic agents and evaluate them for personalized oncology.
Collapse
Affiliation(s)
- Colin C Collins
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tran DD, Corsa CAS, Biswas H, Aft RL, Longmore GD. Temporal and spatial cooperation of Snail1 and Twist1 during epithelial-mesenchymal transition predicts for human breast cancer recurrence. Mol Cancer Res 2011; 9:1644-57. [PMID: 22006115 PMCID: PMC4922748 DOI: 10.1158/1541-7786.mcr-11-0371] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a normal developmental program that is considered to also play an important role in cancer metastasis. Ultimate inducers of EMT are transcriptional repressors that individually can induce experimental EMT, yet in many cells, particularly cancer cells, multiple inducers are expressed simultaneously. Why, and if, and how they interact to regulate EMT is unanswered. Using RNA interference technology to affect protein knockdown and avoid potential overexpression artifact coupled with transient TGFβ treatment to better mimic in vivo conditions we show, in both nontumorigenic and tumorigenic epithelial cancer cells, that Snail1 is uniquely required for EMT initiation, whereas Twist1 is required to maintain late EMT. Twist1, present in resting epithelial cells, is dispensable for EMT initiation. Mechanistically, in response to transient TGFβ treatment, transient Snail1 expression represses Twist1 transcription directly, which is subsequently upregulated, as Snail1 levels decrease, to sustain E-cadherin downregulation and growth arrest of EMT. Persistent Twist1 expression is associated with a p38 and extracellular signal-regulated kinase signal feedback loop that sustains growth-inhibitory signals characteristic of quiescent micrometastatic tumors. This Snail1-Twist1 temporal and spatial cooperation was also observed in vivo during human breast cancer progression to metastasis. Twist1 level, but not Snail1 level, and Twist1:Snail1 ratio in disseminated micrometastatic bone marrow tumor cells was found to correlate with survival and treatment resistance and is highly predictive of metastatic or recurrent disease.
Collapse
Affiliation(s)
- David D. Tran
- Departments of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
| | - Callie Ann S. Corsa
- Departments of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
- The BRIGHT Institute, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
- Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
| | - Hirak Biswas
- Departments of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
- The BRIGHT Institute, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
- Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
| | - Rebecca L. Aft
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
- John Cochran Veterans Administration Hospital, 915 North Grand Boulevard, Saint Louis, MO 63106, USA
| | - Gregory D. Longmore
- Departments of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
- The BRIGHT Institute, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
- Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
| |
Collapse
|