1
|
Liu X, Wang J, Wu LJ, Trinh B, Tsai RYL. IMPDH Inhibition Decreases TERT Expression and Synergizes the Cytotoxic Effect of Chemotherapeutic Agents in Glioblastoma Cells. Int J Mol Sci 2024; 25:5992. [PMID: 38892179 PMCID: PMC11172490 DOI: 10.3390/ijms25115992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
IMP dehydrogenase (IMPDH) inhibition has emerged as a new target therapy for glioblastoma multiforme (GBM), which remains one of the most refractory tumors to date. TCGA analyses revealed distinct expression profiles of IMPDH isoenzymes in various subtypes of GBM and low-grade glioma (LGG). To dissect the mechanism(s) underlying the anti-tumor effect of IMPDH inhibition in adult GBM, we investigated how mycophenolic acid (MPA, an IMPDH inhibitor) treatment affected key oncogenic drivers in glioblastoma cells. Our results showed that MPA decreased the expression of telomerase reverse transcriptase (TERT) in both U87 and U251 cells, and the expression of O6-methylguanine-DNA methyltransferase (MGMT) in U251 cells. In support, MPA treatment reduced the amount of telomere repeats in U87 and U251 cells. TERT downregulation by MPA was associated with a significant decrease in c-Myc (a TERT transcription activator) in U87 but not U251 cells, and a dose-dependent increase in p53 and CCCTC-binding factor (CTCF) (TERT repressors) in both U87 and U251 cells. In U251 cells, MPA displayed strong cytotoxic synergy with BCNU and moderate synergy with irinotecan, oxaliplatin, paclitaxel, or temozolomide (TMZ). In U87 cells, MPA displayed strong cytotoxic synergy with all except TMZ, acting primarily through the apoptotic pathway. Our work expands the mechanistic potential of IMPDH inhibition to TERT/telomere regulation and reveals a synthetic lethality between MPA and anti-GBM drugs.
Collapse
Affiliation(s)
- Xiaoqin Liu
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA; (X.L.); (J.W.); (L.J.W.); (B.T.)
| | - Junying Wang
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA; (X.L.); (J.W.); (L.J.W.); (B.T.)
| | - Laura J. Wu
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA; (X.L.); (J.W.); (L.J.W.); (B.T.)
| | - Britni Trinh
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA; (X.L.); (J.W.); (L.J.W.); (B.T.)
| | - Robert Y. L. Tsai
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA; (X.L.); (J.W.); (L.J.W.); (B.T.)
- Department of Translational Medical Sciences, College of Medicine, Texas A&M University Health Science Center, Houston, TX 77030, USA
| |
Collapse
|
2
|
Camici M, Garcia-Gil M, Pesi R, Allegrini S, Tozzi MG. Purine-Metabolising Enzymes and Apoptosis in Cancer. Cancers (Basel) 2019; 11:cancers11091354. [PMID: 31547393 PMCID: PMC6769685 DOI: 10.3390/cancers11091354] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/03/2019] [Accepted: 09/07/2019] [Indexed: 12/17/2022] Open
Abstract
The enzymes of both de novo and salvage pathways for purine nucleotide synthesis are regulated to meet the demand of nucleic acid precursors during proliferation. Among them, the salvage pathway enzymes seem to play the key role in replenishing the purine pool in dividing and tumour cells that require a greater amount of nucleotides. An imbalance in the purine pools is fundamental not only for preventing cell proliferation, but also, in many cases, to promote apoptosis. It is known that tumour cells harbour several mutations that might lead to defective apoptosis-inducing pathways, and this is probably at the basis of the initial expansion of the population of neoplastic cells. Therefore, knowledge of the molecular mechanisms that lead to apoptosis of tumoural cells is key to predicting the possible success of a drug treatment and planning more effective and focused therapies. In this review, we describe how the modulation of enzymes involved in purine metabolism in tumour cells may affect the apoptotic programme. The enzymes discussed are: ectosolic and cytosolic 5'-nucleotidases, purine nucleoside phosphorylase, adenosine deaminase, hypoxanthine-guanine phosphoribosyltransferase, and inosine-5'-monophosphate dehydrogenase, as well as recently described enzymes particularly expressed in tumour cells, such as deoxynucleoside triphosphate triphosphohydrolase and 7,8-dihydro-8-oxoguanine triphosphatase.
Collapse
Affiliation(s)
- Marcella Camici
- Dipartimento di Biologia, Unità di Biochimica, Via S. Zeno 51, 56127 Pisa, Italy.
| | - Mercedes Garcia-Gil
- Dipartimento di Biologia, Unità di Fisiologia Generale, Via S. Zeno 31, 56127 Pisa, Italy
| | - Rossana Pesi
- Dipartimento di Biologia, Unità di Biochimica, Via S. Zeno 51, 56127 Pisa, Italy
| | - Simone Allegrini
- Dipartimento di Biologia, Unità di Biochimica, Via S. Zeno 51, 56127 Pisa, Italy
| | - Maria Grazia Tozzi
- Dipartimento di Biologia, Unità di Biochimica, Via S. Zeno 51, 56127 Pisa, Italy
| |
Collapse
|
3
|
Kofuji S, Hirayama A, Eberhardt AO, Kawaguchi R, Sugiura Y, Sampetrean O, Ikeda Y, Warren M, Sakamoto N, Kitahara S, Yoshino H, Yamashita D, Sumita K, Wolfe K, Lange L, Ikeda S, Shimada H, Minami N, Malhotra A, Morioka S, Ban Y, Asano M, Flanary VL, Ramkissoon A, Chow LML, Kiyokawa J, Mashimo T, Lucey G, Mareninov S, Ozawa T, Onishi N, Okumura K, Terakawa J, Daikoku T, Wise-Draper T, Majd N, Kofuji K, Sasaki M, Mori M, Kanemura Y, Smith EP, Anastasiou D, Wakimoto H, Holland EC, Yong WH, Horbinski C, Nakano I, DeBerardinis RJ, Bachoo RM, Mischel PS, Yasui W, Suematsu M, Saya H, Soga T, Grummt I, Bierhoff H, Sasaki AT. IMP dehydrogenase-2 drives aberrant nucleolar activity and promotes tumorigenesis in glioblastoma. Nat Cell Biol 2019; 21:1003-1014. [PMID: 31371825 PMCID: PMC6686884 DOI: 10.1038/s41556-019-0363-9] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 06/18/2019] [Indexed: 12/17/2022]
Abstract
In many cancers, high proliferation rates correlate with elevation of rRNA and tRNA levels, and nucleolar hypertrophy. However, the underlying mechanisms linking increased nucleolar transcription and tumorigenesis are only minimally understood. Here we show that IMP dehydrogenase-2 (IMPDH2), the rate-limiting enzyme for de novo guanine nucleotide biosynthesis, is overexpressed in the highly lethal brain cancer glioblastoma. This leads to increased rRNA and tRNA synthesis, stabilization of the nucleolar GTP-binding protein nucleostemin, and enlarged, malformed nucleoli. Pharmacological or genetic inactivation of IMPDH2 in glioblastoma reverses these effects and inhibits cell proliferation, whereas untransformed glia cells are unaffected by similar IMPDH2 perturbations. Impairment of IMPDH2 activity triggers nucleolar stress and growth arrest of glioblastoma cells even in the absence of functional p53. Our results reveal that upregulation of IMPDH2 is a prerequisite for the occurance of aberrant nucleolar function and increased anabolic processes in glioblastoma, which constitutes a primary event in gliomagenesis.
Collapse
Affiliation(s)
- Satoshi Kofuji
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Alexander Otto Eberhardt
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena, Jena, Germany
- Leibniz-Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Risa Kawaguchi
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Oltea Sampetrean
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Yoshiki Ikeda
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mikako Warren
- Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Naoya Sakamoto
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shuji Kitahara
- Department of Anatomy and Developmental Biology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Hirofumi Yoshino
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Daisuke Yamashita
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kazutaka Sumita
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kara Wolfe
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lisa Lange
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena, Jena, Germany
- Leibniz-Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Satsuki Ikeda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Hiroko Shimada
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Noriaki Minami
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Akshiv Malhotra
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Shin Morioka
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuki Ban
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Maya Asano
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Victoria L Flanary
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Annmarie Ramkissoon
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Lionel M L Chow
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Juri Kiyokawa
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tomoyuki Mashimo
- Department of Internal Medicine; Harold C. Simmons Comprehensive Cancer Center; Annette G. Strauss Center for Neuro-Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Greg Lucey
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Sergey Mareninov
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Tatsuya Ozawa
- Division of Human Biology, Solid Tumor and Translational Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Nobuyuki Onishi
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Koichi Okumura
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jumpei Terakawa
- Division of Transgenic Animal Science, Advanced Science Research Center, Kanazawa University, Kanazawa, Japan
| | - Takiko Daikoku
- Division of Transgenic Animal Science, Advanced Science Research Center, Kanazawa University, Kanazawa, Japan
| | - Trisha Wise-Draper
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Nazanin Majd
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kaori Kofuji
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mika Sasaki
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Masaru Mori
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Yonehiro Kanemura
- Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Eric P Smith
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Hiroaki Wakimoto
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Eric C Holland
- Division of Human Biology, Solid Tumor and Translational Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - William H Yong
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Craig Horbinski
- Department of Pathology, University of Kentucky College of Medicine, Lexington, KY, USA
- Departments of Pathology and Neurosurgery, Northwestern University, Chicago, IL, USA
| | - Ichiro Nakano
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ralph J DeBerardinis
- Howard Hughes Medical Institute; Children's Medical Center Research Institute; Department of Pediatrics and Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Robert M Bachoo
- Department of Internal Medicine; Harold C. Simmons Comprehensive Cancer Center; Annette G. Strauss Center for Neuro-Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Paul S Mischel
- Ludwig Institute for Cancer Research; Department of Pathology; Moores Cancer Center, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Wataru Yasui
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
- AMED-CREST, AMED, Tokyo, Japan
| | - Ingrid Grummt
- Division of Molecular Biology of the Cell II, German Cancer Research Center, DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Holger Bierhoff
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena, Jena, Germany
- Leibniz-Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Atsuo T Sasaki
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan.
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Department of Neurosurgery, Brain Tumor Center at UC Gardner Neuroscience Institute, Cincinnati, OH, USA.
| |
Collapse
|
4
|
Bianchi-Smiraglia A, Rana MS, Foley CE, Paul LM, Lipchick BC, Moparthy S, Moparthy K, Fink EE, Bagati A, Hurley E, Affronti HC, Bakin AV, Kandel ES, Smiraglia DJ, Feltri ML, Sousa R, Nikiforov MA. Internally ratiometric fluorescent sensors for evaluation of intracellular GTP levels and distribution. Nat Methods 2017; 14:1003-1009. [PMID: 28869758 PMCID: PMC5636219 DOI: 10.1038/nmeth.4404] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 07/25/2017] [Indexed: 02/07/2023]
Abstract
GTP is a major regulator of multiple cellular processes, but tools for quantitative evaluation of GTP levels in live cells have not been available. We report the development and characterization of genetically encoded GTP sensors, which we constructed by inserting a circularly permuted yellow fluorescent protein (cpYFP) into a region of the bacterial G protein FeoB that undergoes a GTP-driven conformational change. GTP binding to these sensors results in a ratiometric change in their fluorescence, thereby providing an internally normalized response to changes in GTP levels while minimally perturbing those levels. Mutations introduced into FeoB to alter its affinity for GTP created a series of sensors with a wide dynamic range. Critically, in mammalian cells the sensors showed consistent changes in ratiometric signal upon depletion or restoration of GTP pools. We show that these GTP evaluators (GEVALs) are suitable for detection of spatiotemporal changes in GTP levels in living cells and for high-throughput screening of molecules that modulate GTP levels.
Collapse
Affiliation(s)
- Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Mitra S. Rana
- Department of Biochemistry and Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Colleen E. Foley
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Leslie M. Paul
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Brittany C. Lipchick
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Sudha Moparthy
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Kalyana Moparthy
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Emily E. Fink
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Archis Bagati
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Edward Hurley
- Department of Biochemistry and Neurology, Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Hayley C. Affronti
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Andrei V. Bakin
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Eugene S. Kandel
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Dominic J. Smiraglia
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Maria Laura Feltri
- Department of Biochemistry and Neurology, Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Rui Sousa
- Department of Biochemistry and Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Mikhail A. Nikiforov
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| |
Collapse
|
5
|
Targeting MYC Dependence by Metabolic Inhibitors in Cancer. Genes (Basel) 2017; 8:genes8040114. [PMID: 28362357 PMCID: PMC5406861 DOI: 10.3390/genes8040114] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 12/11/2022] Open
Abstract
MYC is a critical growth regulatory gene that is commonly overexpressed in a wide range of cancers. Therapeutic targeting of MYC transcriptional activity has long been a goal, but it has been difficult to achieve with drugs that directly block its DNA-binding ability. Additional approaches that exploit oncogene addiction are promising strategies against MYC-driven cancers. Also, drugs that target metabolic regulatory pathways and enzymes have potential for indirectly reducing MYC levels. Glucose metabolism and oxidative phosphorylation, which can be targeted by multiple agents, promote cell growth and MYC expression. Likewise, modulation of the signaling pathways and protein synthesis regulated by adenosine monophosphate-activated protein kinase (AMPK) and mechanistic target of rapamycin (mTOR) can also be an effective route for suppressing MYC translation. Furthermore, recent data suggest that metabolism of nucleotides, fatty acids and glutamine are exploited to alter MYC levels. Combination therapies offer potential new approaches to overcome metabolic plasticity caused by single agents. Although potential toxicities must be carefully controlled, new inhibitors currently being tested in clinical trials offer significant promise. Therefore, as both a downstream target of metabolism and an upstream regulator, MYC is a prominent central regulator of cancer metabolism. Exploiting metabolic vulnerabilities of MYC-driven cancers is an emerging research area with translational potential.
Collapse
|
6
|
Nguyen D, Liao W, Zeng SX, Lu H. Reviving the guardian of the genome: Small molecule activators of p53. Pharmacol Ther 2017; 178:92-108. [PMID: 28351719 DOI: 10.1016/j.pharmthera.2017.03.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 03/20/2017] [Indexed: 02/07/2023]
Abstract
The tumor suppressor p53 is one of the most important proteins for protection of genomic stability and cancer prevention. Cancers often inactivate it by either mutating its gene or disabling its function. Thus, activating p53 becomes an attractive approach for the development of molecule-based anti-cancer therapy. The past decade and half have witnessed tremendous progress in this area. This essay offers readers with a grand review on this progress with updated information about small molecule activators of p53 either still at bench work or in clinical trials.
Collapse
Affiliation(s)
- Daniel Nguyen
- Department of Biochemistry and Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, 1430 Tulane Ave, LA 70012, United States
| | - Wenjuan Liao
- Department of Biochemistry and Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, 1430 Tulane Ave, LA 70012, United States
| | - Shelya X Zeng
- Department of Biochemistry and Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, 1430 Tulane Ave, LA 70012, United States
| | - Hua Lu
- Department of Biochemistry and Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, 1430 Tulane Ave, LA 70012, United States.
| |
Collapse
|
7
|
Autophagy suppression sensitizes glioma cells to IMP dehydrogenase inhibition-induced apoptotic death. Exp Cell Res 2016; 350:32-40. [PMID: 27818246 DOI: 10.1016/j.yexcr.2016.11.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 09/12/2016] [Accepted: 11/02/2016] [Indexed: 11/24/2022]
Abstract
We investigated the role of autophagy, a process of controlled self-digestion, in the in vitro anticancer action of the inosine monophosphate dehydrogenase (IMPDH) inhibitor ribavirin. Ribavirin-triggered oxidative stress, caspase activation, and apoptotic death in U251 human glioma cells were associated with the induction of autophagy, as confirmed by intracellular acidification, appearance of autophagic vesicles, conversion of microtubule associated protein 1 light chain 3 (LC3)-I to autophagosome-associated LC3-II, and degradation of autophagic target p62/sequestosome 1. Ribavirin downregulated the activity of autophagy-inhibiting mammalian target of rapamycin complex 1 (mTORC1), as indicated by a decrease in phosphorylation of the mTORC1 substrate ribosomal p70S6 kinase and reduction of the mTORC1-activating Src/Akt signaling. Guanosine supplementation inhibited, while IMPDH inhibitor tiazofurin mimicked ribavirin-mediated autophagy induction, suggesting the involvement of IMPDH blockade in the observed effect. Autophagy suppression by ammonium chloride, bafilomycin A1, or RNA interference-mediated knockdown of LC3 sensitized glioma cells to ribavirin-induced apoptosis. Ribavirin also induced cytoprotective autophagy associated with Akt/mTORC1 inhibition in C6 rat glioma cells. Our data demonstrate that ribavirin-triggered Akt/mTORC1-dependent autophagy counteracts apoptotic death of glioma cells, indicating autophagy suppression as a plausible therapeutic strategy for sensitization of cancer cells to IMPDH inhibition.
Collapse
|
8
|
Meshkini A. Fine-tuning of the cellular signaling pathways by intracellular GTP levels. Cell Biochem Biophys 2015; 70:27-32. [PMID: 24643502 DOI: 10.1007/s12013-014-9897-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
It has become increasingly evident that among purine nucleotides, guanine based nucleotides specially guanosine-5'-triphosphate (GTP) serve as an important and independent regulatory factors for development and diverse cellular functions such as differentiation, metabolism, proliferation and survival in multiple tissues. In this brief review, it has been provided selective outline related to delicate regulation of signaling pathways by guanosine based nucleotides as intracellular signaling molecules. Although the exact mode of action of theses nucleotides in many biological processes and signaling pathways is still elusive, it has become well clear that intracellular guanosine based nucleotides content rather than adenosine based nucleotides could modulate the intensity and duration of signaling which ultimately impact on cell's fate. It opens an entirely new perspective for developing new and potential therapeutic strategies to combat diseases like cancer, hypoxia, etc.
Collapse
Affiliation(s)
- Azadeh Meshkini
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran,
| |
Collapse
|
9
|
Cho SY, Lee JH, Ju MK, Jeong EM, Kim HJ, Lim J, Lee S, Cho NH, Park HH, Choi K, Jeon JH, Kim IG. Cystamine induces AIF-mediated apoptosis through glutathione depletion. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:619-31. [PMID: 25549939 DOI: 10.1016/j.bbamcr.2014.12.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 12/09/2014] [Accepted: 12/22/2014] [Indexed: 12/18/2022]
Abstract
Cystamine and its reduced form cysteamine showed protective effects in various models of neurodegenerative disease, including Huntington's disease and Parkinson's disease. Other lines of evidence demonstrated the cytotoxic effect of cysteamine on duodenal mucosa leading to ulcer development. However, the mechanism for cystamine cytotoxicity remains poorly understood. Here, we report a new pathway in which cystamine induces apoptosis by targeting apoptosis-inducing factor (AIF). By screening of various cell lines, we observed that cystamine and cysteamine induce cell death in a cell type-specific manner. Comparison between cystamine-sensitive and cystamine-resistant cell lines revealed that cystamine cytotoxicity is not associated with unfolded protein response, reactive oxygen species generation and transglutaminase or caspase activity; rather, it is associated with the ability of cystamine to trigger AIF nuclear translocation. In cystamine-sensitive cells, cystamine suppresses the levels of intracellular glutathione by inhibiting γ-glutamylcysteine synthetase expression that triggers AIF translocation. Conversely, glutathione supplementation completely prevents cystamine-induced AIF translocation and apoptosis. In rats, cysteamine administration induces glutathione depletion and AIF translocation leading to apoptosis of duodenal epithelium. These results indicate that AIF translocation through glutathione depletion is the molecular mechanism of cystamine toxicity, and provide important implications for cystamine in the neurodegenerative disease therapeutics as well as in the regulation of AIF-mediated cell death.
Collapse
Affiliation(s)
- Sung-Yup Cho
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Jin-Haeng Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Mi-kyeong Ju
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Eui Man Jeong
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Hyo-Jun Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Jisun Lim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Seungun Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Nam-Hyuk Cho
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Hyun Ho Park
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan 712-749, Republic of Korea
| | - Kihang Choi
- Department of Chemistry, Korea University, Seoul 136-701, Republic of Korea
| | - Ju-Hong Jeon
- Institute of Human-Environment Interface Biology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - In-Gyu Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea.
| |
Collapse
|
10
|
Zhang Q, Zhou X, Wu R, Mosley A, Zeng SX, Xing Z, Lu H. The role of IMP dehydrogenase 2 in Inauhzin-induced ribosomal stress. eLife 2014; 3. [PMID: 25347121 PMCID: PMC4209374 DOI: 10.7554/elife.03077] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 10/06/2014] [Indexed: 11/13/2022] Open
Abstract
The 'ribosomal stress (RS)-p53 pathway' is triggered by any stressor or genetic alteration that disrupts ribosomal biogenesis, and mediated by several ribosomal proteins (RPs), such as RPL11 and RPL5, which inhibit MDM2 and activate p53. Inosine monophosphate (IMP) dehydrogenase 2 (IMPDH2) is a rate-limiting enzyme in de novo guanine nucleotide biosynthesis and crucial for maintaining cellular guanine deoxy- and ribonucleotide pools needed for DNA and RNA synthesis. It is highly expressed in many malignancies. We previously showed that inhibition of IMPDH2 leads to p53 activation by causing RS. Surprisingly, our current study reveals that Inauzhin (INZ), a novel non-genotoxic p53 activator by inhibiting SIRT1, can also inhibit cellular IMPDH2 activity, and reduce the levels of cellular GTP and GTP-binding nucleostemin that is essential for rRNA processing. Consequently, INZ induces RS and the RPL11/RPL5-MDM2 interaction, activating p53. These results support the new notion that INZ suppresses cancer cell growth by dually targeting SIRT1 and IMPDH2.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, United States
| | - Xiang Zhou
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, United States
| | - RuiZhi Wu
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, United States
| | - Amber Mosley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, United States
| | - Shelya X Zeng
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, United States
| | - Zhen Xing
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, United States
| | - Hua Lu
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, United States
| |
Collapse
|
11
|
Wawrzyniak JA, Bianchi-Smiraglia A, Bshara W, Mannava S, Ackroyd J, Bagati A, Omilian AR, Im M, Fedtsova N, Miecznikowski JC, Moparthy KC, Zucker SN, Zhu Q, Kozlova NI, Berman AE, Hoek KS, Gudkov AV, Shewach DS, Morrison CD, Nikiforov MA. A purine nucleotide biosynthesis enzyme guanosine monophosphate reductase is a suppressor of melanoma invasion. Cell Rep 2013; 5:493-507. [PMID: 24139804 DOI: 10.1016/j.celrep.2013.09.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 08/20/2013] [Accepted: 09/11/2013] [Indexed: 01/02/2023] Open
Abstract
Melanoma is one of the most aggressive types of human cancers, and the mechanisms underlying melanoma invasive phenotype are not completely understood. Here, we report that expression of guanosine monophosphate reductase (GMPR), an enzyme involved in de novo biosynthesis of purine nucleotides, was downregulated in the invasive stages of human melanoma. Loss- and gain-of-function experiments revealed that GMPR downregulates the amounts of several GTP-bound (active) Rho-GTPases and suppresses the ability of melanoma cells to form invadopodia, degrade extracellular matrix, invade in vitro, and grow as tumor xenografts in vivo. Mechanistically, we demonstrated that GMPR partially depletes intracellular GTP pools. Pharmacological inhibition of de novo GTP biosynthesis suppressed whereas addition of exogenous guanosine increased invasion of melanoma cells as well as cells from other cancer types. Our data identify GMPR as a melanoma invasion suppressor and establish a link between guanosine metabolism and Rho-GTPase-dependent melanoma cell invasion.
Collapse
Affiliation(s)
- Joseph A Wawrzyniak
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Huh KH, Cho Y, Kim BS, Do JH, Park YJ, Joo DJ, Kim MS, Kim YS. The role of thioredoxin 1 in the mycophenolic acid-induced apoptosis of insulin-producing cells. Cell Death Dis 2013; 4:e721. [PMID: 23846223 PMCID: PMC3730420 DOI: 10.1038/cddis.2013.247] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 06/07/2013] [Accepted: 06/07/2013] [Indexed: 01/12/2023]
Abstract
Mycophenolic acid (MPA) is one of many effective immunosuppressive drugs. However, MPA can induce cellular toxicity and impair cellular function in β-cells. To explore the effects of MPA and the relation between MPA and Trx-1, we used various methods, including an Illumina microarray, to identify the genes regulated during pancreatic β-cell death following MPA treatment. INS-1E cells (a pancreatic β-cell line) and isolated rat islets were treated with MPA for 12, 24, or 36 h, and subsequent microarray analysis showed that (Trx1) gene expression was significantly reduced by MPA. Further, Trx1 overexpression increased the cell viability, decreased the activations of c-jun N-terminal kinase (JNK) and caspase-3 by MPA, and attenuated ROS upregulation by MPA. Furthermore, siRNA knockdown of Trx1 increased MPA-induced cell death and the activations of p-JNK and caspase-3, and MPA significantly provoked the apoptosis of insulin-secreting cells via Trx1 downregulation. Our findings suggest that the prevention of Trx1 downregulation in response to MPA is critical for successful islet transplantation.
Collapse
Affiliation(s)
- K H Huh
- Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
13
|
|
14
|
Lin T, Meng L, Tsai RYL. GTP depletion synergizes the anti-proliferative activity of chemotherapeutic agents in a cell type-dependent manner. Biochem Biophys Res Commun 2011; 414:403-8. [PMID: 21971546 DOI: 10.1016/j.bbrc.2011.09.091] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 09/18/2011] [Indexed: 10/17/2022]
Abstract
Mycophenolic acid (MPA) depletes intracellular GTP by blocking de novo guanine nucleotide synthesis. GTP is used ubiquitously for DNA/RNA synthesis and as a signaling molecule. Here, we made a surprising discovery that the anti-proliferative activity of MPA acts synergistically with specific chemotherapeutic agents in a cell type-dependent manner. In MDA-MB-231 cells, MPA shows an extremely potent synergy with 5-FU but not with doxorubicin or etoposide. The synergy between 5-FU and MPA works most effectively against the highly tumorigenic mammary tumor cells compared to the less tumorigenic ones, and does not work in the non-breast cancer cell types that we tested, with the exception of PC3 cells. On the contrary, MPA shows the highest synergy with paclitaxel but not with 5-FU in SCC-25 cells, derived from oral squamous cell carcinomas. Mechanistically, the synergistic effect of MPA on 5-FU in MDA-MB-231 cells can be recapitulated by inhibiting the RNA polymerase-I activity and requires the expression of nucleostemin. This work reveals that the synergy between MPA and anti-proliferative agents is determined by cell type-dependent factors.
Collapse
Affiliation(s)
- Tao Lin
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA
| | | | | |
Collapse
|
15
|
Nouri K, Yazdanparast R. Effects of 3-Hydrogenkwadaphnin on intracellular purine nucleotide contents and their link to K562 cell death. Food Chem 2011; 128:81-6. [DOI: 10.1016/j.foodchem.2011.02.080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 12/22/2010] [Accepted: 02/24/2011] [Indexed: 11/15/2022]
|
16
|
Intracellular GTP level determines cell's fate toward differentiation and apoptosis. Toxicol Appl Pharmacol 2011; 253:188-96. [PMID: 21396949 DOI: 10.1016/j.taap.2011.02.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 02/09/2011] [Accepted: 02/28/2011] [Indexed: 01/23/2023]
Abstract
Since the adequate supply of guanine nucleotides is vital for cellular activities, limitation of their syntheses would certainly result in modulation of cellular fate toward differentiation and apoptosis. The aim of this study was to set a correlation between the intracellular level of GTP and the induction of relevant signaling pathways involved in the cell's fate toward life or death. In that regard, we measured the GTP level among human leukemia K562 cells exposed to mycophenolic acid (MPA) or 3-hydrogenkwadaphnin (3-HK) as two potent inosine monophosphate dehydrogenase inhibitors. Our results supported the maturation of the cells when the intracellular GTP level was reduced by almost 30-40%. Under these conditions, 3-HK and/or MPA caused up-regulation of PKCα and PI3K/AKT pathways. Furthermore, co-treatment of cells with hypoxanthine plus 3-HK or MPA, which caused a reduction of about 60% in the intracellular GTP levels, led to apoptosis and activation of mitochondrial pathways through inverse regulation of Bcl-2/Bax expression and activation of caspase-3. Moreover, our results demonstrated that attenuation of GTP by almost 60% augmented the intracellular ROS and nuclear localization of p21 and subsequently led to cell death. These results suggest that two different threshold levels of GTP are needed for induction of differentiation and/or ROS-associated apoptosis.
Collapse
|
17
|
He X, Smeets RL, Koenen HJPM, Vink PM, Wagenaars J, Boots AMH, Joosten I. Mycophenolic acid-mediated suppression of human CD4+ T cells: more than mere guanine nucleotide deprivation. Am J Transplant 2011; 11:439-49. [PMID: 21342445 DOI: 10.1111/j.1600-6143.2010.03413.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Mycophenolic acid is the active ingredient of the immunosuppressant mycophenolate mofetil that is widely used in transplantation medicine and autoimmunity. Mycophenolic acid inhibits inosine monophosphate dehydrogenase, an enzyme involved in biosynthesis of guanine nucleotides required for lymphocyte clonal expansion. Here, we present novel insights into the mechanisms underlying mycophenolic acid-mediated suppression of human CD4+ T cells. Upon CD3/CD28 stimulation, mycophenolic acid inhibited T cell IL-17, IFN-γ and TNF-α production but not IL-2 production. Phenotypic analysis showed that drug treatment enhanced the expression of negative co-stimulators PD-1, CTLA-4 and the transcription factor FoxP3 and decreased the expression of positive co-stimulators CD27 and CD28, whereas CD25 was unaffected. Mycophenolic acid-treated cells were anergic, but not suppressive, and at the same time proved hyperblastoid with high metabolic activity. Moreover, a reduced Akt/mTOR and STAT5 signaling was observed. Interestingly, the co-stimulatory molecule CD70 was uniquely and dose-dependently upregulated on mycophenolic acid-treated T cells and found to be directly linked to target enzyme inhibition. CD70 on mycophenolic acid-treated cells proved functional: an anti-CD70 agonist was found to restore both STAT5 and Akt/mTOR signaling and may thereby prevent apoptosis and promote survival. These novel insights may contribute to optimization of protocols for MPA-based immunosuppressive regimens.
Collapse
Affiliation(s)
- X He
- Department of Laboratory Medicine, Laboratory Medical Immunology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
18
|
Cho SY, Lee JH, Bae HD, Jeong EM, Jang GY, Kim CW, Shin DM, Jeon JH, Kim IG. Transglutaminase 2 inhibits apoptosis induced by calcium- overload through down-regulation of Bax. Exp Mol Med 2011; 42:639-50. [PMID: 20676023 DOI: 10.3858/emm.2010.42.9.063] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
An abrupt increase of intracellular Ca(2+) is observed in cells under hypoxic or oxidatively stressed conditions. The dysregulated increase of cytosolic Ca(2+) triggers apoptotic cell death through mitochondrial swelling and activation of Ca(2+)-dependent enzymes. Transglutaminase 2 (TG2) is a Ca(2+)-dependent enzyme that catalyzes transamidation reaction producing cross-linked and polyaminated proteins. TG2 activity is known to be involved in the apoptotic process. However, the pro-apoptotic role of TG2 is still controversial. In this study, we investigate the role of TG2 in apoptosis induced by Ca(2+)-overload. Overexpression of TG2 inhibited the A23187-induced apoptosis through suppression of caspase-3 and -9 activities, cytochrome c release into cytosol, and mitochondria membrane depolarization. Conversely, down-regulation of TG2 caused the increases of cell death, caspase-3 activity and cytochrome c in cytosol in response to Ca(2+)-overload. Western blot analysis of Bcl-2 family proteins showed that TG2 reduced the expression level of Bax protein. Moreover, overexpression of Bax abrogated the anti-apoptotic effect of TG2, indicating that TG2-mediated suppression of Bax is responsible for inhibiting cell death under Ca(2+)-overloaded conditions. Our findings revealed a novel anti-apoptotic pathway involving TG2, and suggested the induction of TG2 as a novel strategy for promoting cell survival in diseases such as ischemia and neurodegeneration.
Collapse
Affiliation(s)
- Sung-Yup Cho
- Department of Biochemistry and Molecular Biology/Aging and Apoptosis Research Center (AARC), Seoul National University College of Medicine, Seoul 110-799, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Zhao M, Qu H. Human liver rate-limiting enzymes influence metabolic flux via branch points and inhibitors. BMC Genomics 2009; 10 Suppl 3:S31. [PMID: 19958496 PMCID: PMC2788385 DOI: 10.1186/1471-2164-10-s3-s31] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background Rate-limiting enzymes, because of their relatively low velocity, are believed to influence metabolic flux in pathways. To investigate their regulatory role in metabolic networks, we look at the global organization and interactions between rate-limiting enzymes and compounds such as branch point metabolites and enzyme inhibitors in human liver. Results Based on 96 rate-limiting enzymes and 132 branch point compounds from human liver, we found that rate-limiting enzymes surrounded 76.5% of branch points. In a compound conversion network from human liver, the 128 branch points involved showed a dramatically higher average degree, betweenness centrality and closeness centrality as a whole. Nearly half of the in vivo inhibitors were products of rate-limiting enzymes, and covered 75.34% of the inhibited targets in metabolic inhibitory networks. Conclusion From global topological organization, rate-limiting enzymes as a whole surround most of the branch points; so they can influence the flux through branch points. Since nearly half of the in vivo enzyme inhibitors are produced by rate-limiting enzymes in human liver, these enzymes can initiate inhibitory regulation and then influence metabolic flux through their natural products.
Collapse
Affiliation(s)
- Min Zhao
- Center for Bioinformatics, National Laboratory of Protein Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing, 100871, PR China.
| | | |
Collapse
|
20
|
Liu YX, Jin LM, Zhou L, Xie HY, Jiang GP, Wang Y, Feng XW, Chen H, Yan S, Zheng SS. Mycophenolate mofetil attenuates liver ischemia/reperfusion injury in rats. Transpl Int 2009; 22:747-56. [DOI: 10.1111/j.1432-2277.2009.00866.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
21
|
Faugaret D, Dauba A, Baron C, Velge-Roussel F, Lebranchu Y. Mycophenolic Acid Inhibits p38 Mitogen-Activated Protein Kinase in Human Monocyte-Derived Dendritic Cells Stimulated by Lipopolysaccharide. Transplant Proc 2009; 41:698-9. [PMID: 19328959 DOI: 10.1016/j.transproceed.2008.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dendritic cell (DC) maturation, a crucial stage in the immune response, can be induced by various stimuli, such as lipopolysaccharide (LPS). Maturation signals trigger up-regulation of costimulatory molecule expression, increasing the ability of DCs to prime T helper cells. We and others have previously reported that mycophenolic acid (MPA) inhibits DC maturation and activation. However, the mechanisms remain unknown. The primary effect of MPA is inhibition of inosine monophosphate dehydrogenase (IMPDH), an enzyme involved in the de novo synthesis of guanosine nucleotide. The process of DC maturation is highly dependent on mitogen-activated protein kinase (MAPK) phosphorylation, especially p38MAPK. We therefore decided to study whether MPA affects these processes. Human monocyte-derived DCs were activated by LPS in the presence or absence of MPA. To assess whether the depletion of guanine affected p38MAPK phosphorylation, increasing doses of exogenous guanosine were added before stimulation. The results by flow cytometry showed that MPA inhibited p38MAPK phosphorylation by 25%. Interestingly, exogenous guanosine did not reverse the MPA inhibition. Our results suggested that MPA inhibits p38MAPK activity independent of IMPDH in human DCs. This effect of MPA may explain its capacity to inhibit maturation marker expression on DCs.
Collapse
Affiliation(s)
- D Faugaret
- Université François-Rabelais, EA 4245 Cellules Dendritiques et Greffes, IFR 136, UFR de Médecine, Tours, France
| | | | | | | | | |
Collapse
|
22
|
The RhoGDI-α/JNK signaling pathway plays a significant role in mycophenolic acid-induced apoptosis in an insulin-secreting cell line. Cell Signal 2009; 21:356-64. [DOI: 10.1016/j.cellsig.2008.11.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2008] [Revised: 11/12/2008] [Accepted: 11/12/2008] [Indexed: 11/23/2022]
|
23
|
Chan ER, Lavender H, Li G, Haviernik P, Bunting KD, Adams MD. An ENU-induced recessive mutation in Mpl leads to thrombocytopenia with overdominance. Exp Hematol 2008; 37:276-84. [PMID: 19059699 DOI: 10.1016/j.exphem.2008.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Revised: 10/07/2008] [Accepted: 10/08/2008] [Indexed: 01/07/2023]
Abstract
OBJECTIVE The aim of this study was to identify and characterize the causative mutation in the thrombocytopenic mouse strain HLB219 that was generated at the Jackson Laboratory as part of a large-scale N-ethyl-N-nitrosourea mutagenesis screen. MATERIALS AND METHODS The HLB219 mutation was identified by interval mapping of F2 mice generated from intercross breeding of HLB219 to both BALB/cByJ (BALB) and 129/SvImJ (129/Sv). Mpl was identified as a candidate gene and sequenced. The mutation was characterized in vivo in mouse hematopoietic stem/progenitor cell assays and in cell culture by expression in Ba/F3 cells. RESULTS A novel mutation in the thrombopoietin (TPO) receptor Mpl in HLB219 mice caused a Cys-->Arg substitution at codon 40 in the extracellular region of the receptor. Mice homozygous for the Mpl(hlb219) mutation had an 80% decrease in the number of platelets in comparison to the wild-type C57BL/6J strain, low numbers of bone marrow megakaryocytes, high TPO levels, and decreased competitive repopulating ability, consistent with a loss-of-function mutation in the receptor. Mice heterozygous for Mpl(hlb219) however, showed an overdominance effect with a significant increase in platelet number. Functional analysis in vitro demonstrated that Ba/F3 cells expressing the mutant MPL(hlb219) protein failed to activate extracellular signal-regulated kinase and signal transducers and activators of transcription 5, but proliferated in the absence of TPO and required constitutive phosphorylation of RAC-alpha serine/threonine protein kinase (AKT) for cytokine-independent growth. CONCLUSION Thrombocytopenia in HLB219 mice is caused by a recessive mutation in Mpl that abrogates mitogen-activated protein kinase-extracellular signal regulated kinase and janus kinase-signal transducers and activators of transcription signaling.
Collapse
Affiliation(s)
- E Ricky Chan
- Department of Genetics, Case Western Reserve University, Cleveland, Ohio 44106-4955, USA
| | | | | | | | | | | |
Collapse
|
24
|
Sun XX, Dai MS, Lu H. Mycophenolic acid activation of p53 requires ribosomal proteins L5 and L11. J Biol Chem 2008; 283:12387-92. [PMID: 18305114 DOI: 10.1074/jbc.m801387200] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mycophenolate mofetil (MMF), a prodrug of mycophenolic acid (MPA), is widely used as an immunosuppressive agent. MPA selectively inhibits inosine monophosphate dehydrogenase (IMPDH), a rate-limiting enzyme for the de novo synthesis of guanine nucleotides, leading to depletion of the guanine nucleotide pool. Its chemotherapeutic effects have been attributed to its ability to induce cell cycle arrest and apoptosis. MPA treatment has also been shown to induce and activate p53. However, the mechanism underlying the p53 activation pathway is still unclear. Here, we show that MPA treatment results in inhibition of pre-rRNA synthesis and disruption of the nucleolus. This treatment enhances the interaction of MDM2 with L5 and L11. Interestingly, knockdown of endogenous L5 or L11 markedly impairs the induction of p53 and G(1) cell cycle arrest induced by MPA. These results suggest that MPA may trigger a nucleolar stress that induces p53 activation via inhibition of MDM2 by ribosomal proteins L5 and L11.
Collapse
Affiliation(s)
- Xiao-Xin Sun
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine and Simon Cancer Center, Indianapolis, Indiana 46202, USA
| | | | | |
Collapse
|
25
|
Bolin C, Cardozo-Pelaez F. Assessing biomarkers of oxidative stress: analysis of guanosine and oxidized guanosine nucleotide triphosphates by high performance liquid chromatography with electrochemical detection. J Chromatogr B Analyt Technol Biomed Life Sci 2007; 856:121-30. [PMID: 17581804 PMCID: PMC2072907 DOI: 10.1016/j.jchromb.2007.05.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2007] [Revised: 05/18/2007] [Accepted: 05/21/2007] [Indexed: 12/11/2022]
Abstract
Oxidation of the guanosine moiety in DNA has become a hallmark biomarker in assessing oxidative stress. The oxidation of guanosine in the nucleotide triphosphate pool has been overlooked due to the lack of a reliable methodology. This method describes a sample processing and high performance liquid chromatography with electrochemical detection protocol for the analysis of the cellular pool of guanosine triphosphates and oxidized guanosine triphosphates. Validation of this method is demonstrated along with evaluation of these analytes in control and oxidizing conditions in vitro and in HEK 293T cells. Oxidation of this triphosphate pool occurred independently of oxidation to DNA.
Collapse
|
26
|
Huang M, Ji Y, Itahana K, Zhang Y, Mitchell B. Guanine nucleotide depletion inhibits pre-ribosomal RNA synthesis and causes nucleolar disruption. Leuk Res 2007; 32:131-41. [PMID: 17462731 PMCID: PMC4552191 DOI: 10.1016/j.leukres.2007.03.025] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2007] [Revised: 02/26/2007] [Accepted: 03/12/2007] [Indexed: 12/13/2022]
Abstract
Inosine monophosphate dehydrogenase (IMPDH) is a pivotal enzyme in the de novo pathway of guanine nucleotide biosynthesis. Inhibitors of this enzyme decrease intracellular guanine nucleotide levels by 50-80% and have potential as anti-neoplastic agents. Both mycophenolic acid (MPA) and AVN-944 are highly specific inhibitors of IMPDH that cause cell cycle arrest or apoptosis in lymphocytes and leukemic cell lines. We have examined the mechanisms by which these two agents cause cytotoxicity. Both MPA and AVN-944 inhibit the growth of K562 cells, and induce apoptosis in Raji B and CCRF-CEM T cells. Both compounds strikingly inhibit RNA synthesis within 2 h of exposure. Depletion of guanine nucleotides by MPA and AVN-944 also causes an early and near-complete reduction in levels of the 45S precursor rRNA synthesis and the concomitant translocation of nucleolar proteins including nucleolin, nucleophosmin, and nucleostemin from the nucleolus to the nucleoplasm. This efflux correlates temporally with the sustained induction of p53 in cell lines with wild-type p53. We conclude that inhibition of IMPDH causes a primary reduction in rRNA synthesis and secondary nucleolar disruption and efflux of nucleolar proteins that most likely mediate cell cycle arrest or apoptosis. The ability of AVN-944 to induce apoptosis in a number of leukemic cell lines supports its potential utility in the treatment of hematologic malignancies.
Collapse
Affiliation(s)
- Min Huang
- Department of Medicine, Division of Oncology, Stanford University. Stanford, CA, USA
| | - Yanshan Ji
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Koji Itahana
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yanping Zhang
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Beverly Mitchell
- Department of Medicine, Division of Oncology, Stanford University. Stanford, CA, USA
- Corresponding author: Dr. Beverly S. Mitchell, Department of Medicine, Division of Oncology, Stanford University. Stanford, CA 94305-5796, Telephone: 650-725-9621, Fax: 650-736-0607, Email address:
| |
Collapse
|
27
|
Boratyńska M, Zmonarski SC, Klinger M. Reccurence of Kaposi's sarcoma after increased exposure to sirolimus. Int Immunopharmacol 2006; 6:2018-22. [DOI: 10.1016/j.intimp.2006.09.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Accepted: 09/15/2006] [Indexed: 12/26/2022]
|
28
|
Kim JY, Yoon SY, Park J, Kim YS. Mycophenolic Acid Induces Islet Apoptosis by Regulating Mitogen-Activated Protein Kinase Activation. Transplant Proc 2006; 38:3277-9. [PMID: 17175248 DOI: 10.1016/j.transproceed.2006.10.124] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2006] [Indexed: 11/23/2022]
Abstract
Mycophenolic acid (MPA), an inosine monophosphate dehydrogenase inhibitor, is widely used as an immunosuppressive drug after transplantations including those of pancreas islet cells. However, recent reports have indicated that MPA has apoptotic effects on islet cells in vitro. To study the effect of MPA on islet cells and determine its mechanism, we used an insulin secreting cell-line, HIT-T15. We examined mitogen-activated protein kinase (MAPK) activation after MPA treatment, and determining cell death levels using methylthiazdetetrazolium assays. The activations of extracellular signal-regulated protein kinase (ERK), c-jun N-terminal kinase (JNK), and p38 MAPK and caspase-3 cleavage were measured by Western blotting. MPA (1, 10, 30 micromol/L) increased cell death and caspase-3 cleavage within 24 hours. Exogenous 500 micromol/L guanosine reversed the MPA-induced islet cell death, but exogenous adenosine did not. MPA 10 micromol/L induced cell apoptosis and increased the activations of JNK, ERK, and p38 MAPK. Furthermore, exogenous guanosine, but not exogenous adenosine, reversed these effects induced by MPA. This study demonstrated that MPA may induce islet apoptosis in HIT-T15 cells by increasing activations of JNK, ERK, and p38 MAPK in a guanosine-dependent manner.
Collapse
Affiliation(s)
- J Y Kim
- Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, South Korea
| | | | | | | |
Collapse
|
29
|
Ha H, Kim MS, Park J, Huh JY, Huh KH, Ahn HJ, Kim YS. Mycophenolic acid inhibits mesangial cell activation through p38 MAPK inhibition. Life Sci 2006; 79:1561-7. [PMID: 16740277 DOI: 10.1016/j.lfs.2006.05.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2005] [Revised: 04/25/2006] [Accepted: 05/01/2006] [Indexed: 10/24/2022]
Abstract
Mesangial cell (MC) proliferation and extracellular matrix (ECM) accumulation are major pathologic features of chronic renal disease including chronic allograft nephropathy (CAN). Mycophenolic acid (MPA), a potent immunosuppressant, has emerged as a treatment to prevent CAN because it inhibits MC proliferation and ECM synthesis, but the mechanism involved has not been clarified. The present study examined relative role of extracellular signal-regulated kinase 1/2 (ERK1/2) and p38 mitogen-activated protein kinase (p38 MAPK) activation in inhibitory effect of MPA on MC activation. Growth arrested and synchronized primary rat MC (passages 7-11) were stimulated by PDGF 10 ng/ml in the presence and absence of clinically attainable dose of MPA (0-10 microM). Cell proliferation was assessed by [(3)H]thymidine incorporation, fibronectin and the activation of ERK and p38 MAPK by Western blot analysis, and total collagen by [(3)H]proline incorporation. PDGF increased cell proliferation by 4.6-fold, fibronectin secretion by 3.2-fold, total collagen synthesis by 1.8-fold, and the activation of ERK and 38 MAPK by 5.6-fold and 3.1-fold, respectively, compared to control. MPA, at doses inhibiting PDGF-induced MC proliferation and ECM synthesis, effectively blocked p38 MAPK activation but reduced ERK activation by 23% at maximal concentration tested (10 microM). Exogenous guanosine partially reversed the inhibition of MPA on p38 MAPK activation. Inhibitor of ERK or p38 MAPK suppressed PDGF-induced MC proliferation and ECM synthesis. In conclusion, MPA inhibits p38 MAPK activation leading to inhibiting proliferation and ECM synthesis in MC. Guanosine reduction is partially responsible for inhibitory effect of MPA on p38 MAPK activation in MC.
Collapse
Affiliation(s)
- Hunjoo Ha
- Ewha Womans University College of Pharmacy, Seoul, South Korea
| | | | | | | | | | | | | |
Collapse
|
30
|
Mortimer S, Hedstrom L. Autosomal dominant retinitis pigmentosa mutations in inosine 5'-monophosphate dehydrogenase type I disrupt nucleic acid binding. Biochem J 2005; 390:41-7. [PMID: 15882147 PMCID: PMC1184561 DOI: 10.1042/bj20042051] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Two mutations of IMPDH1 (inosine 5'-monophosphate dehydrogenase type I), R224P and D226N, have recently been found to cause adRP (autosomal dominant retinitis pigmentosa). IMPDH1 catalyses the rate-limiting step in guanine nucleotide biosynthesis and also binds single-stranded nucleic acids. In the present paper, we report the biochemical characterization of the adRP-linked mutations, R224P and D226N, and a potentially pathogenic mutation, V268I. The adRP-linked mutations have no effect on enzyme activity, protein stability or protein aggregation. These results suggest strongly that the mutations do not affect enzyme activity in vivo and thus do not perturb the guanine nucleotide pool. The R224P mutation changes the distribution of enzyme between the nucleus and cytoplasm. This effect was not observed with the D226N mutation, so the relevance of this observation to disease is unclear. In contrast, both mutations decrease the affinity of nucleic acid binding and both fail to co-immunoprecipitate RNA. These observations suggest that nucleic acid binding provides a functional assay for adRP pathogenicity. The putative adRP-linked mutation V268I also disrupts nucleic acid binding, which suggests that this mutation is indeed pathogenic.
Collapse
Affiliation(s)
- Sarah E. Mortimer
- Department of Biochemistry, Brandeis University, 415 South St., Waltham MA 02454-9110, U.S.A
| | - Lizbeth Hedstrom
- Department of Biochemistry, Brandeis University, 415 South St., Waltham MA 02454-9110, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
31
|
Ji Y, Gu J, Makhov AM, Griffith JD, Mitchell BS. Regulation of the interaction of inosine monophosphate dehydrogenase with mycophenolic Acid by GTP. J Biol Chem 2005; 281:206-12. [PMID: 16243838 DOI: 10.1074/jbc.m507056200] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Inosine monophosphate dehydrogenase (IMPDH), a rate-limiting enzyme in the de novo synthesis of guanine nucleotides, is a major therapeutic target. A prototypic uncompetitive inhibitor of IMPDH, mycophenolic acid (MPA), is the active form of mycophenolate mofeteil (CellCept), a widely used immunosuppressive drug. We have found that MPA interacts with intracellular IMPDH in vivo to alter its mobility on SDS-polyacrylamide gels. MPA also induces a striking conformational change in IMPDH protein in intact cells, resulting in the formation of annular aggregates of protein with concomitant inhibition of IMPDH activity. These aggregates are not associated with any known intracellular organelles and are reversible by incubating cells with guanosine, which repletes intracellular GTP, or with GTPgammaS. GTP also restores IMPDH activity. Treatment of highly purified IMPDH with MPA also results in the formation of large aggregates of protein, a process that is both prevented and reversed by the addition of GTP. Finally, GTP binds to IMPDH at physiologic concentrations, induces the formation of linear arrays of tetrameric protein, and prevents the aggregation of protein induced by MPA. We conclude that intracellular GTP acts as an antagonist to MPA by directly binding to IMPDH and reversing the conformational changes in the protein.
Collapse
Affiliation(s)
- YanShan Ji
- Department of Pharmacology and The Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599-7295, USA
| | | | | | | | | |
Collapse
|
32
|
Ishitsuka K, Hideshima T, Hamasaki M, Raje N, Kumar S, Podar K, Le Gouill S, Shiraishi N, Yasui H, Roccaro AM, Tai YZ, Chauhan D, Fram R, Tamura K, Jain J, Anderson KC. Novel inosine monophosphate dehydrogenase inhibitor VX-944 induces apoptosis in multiple myeloma cells primarily via caspase-independent AIF/Endo G pathway. Oncogene 2005; 24:5888-96. [PMID: 15940263 DOI: 10.1038/sj.onc.1208739] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Inosine monophosphate dehydrogenase (IMPDH) is a rate-limiting enzyme required for the de novo synthesis of guanine nucleotides from IMP. VX-944 (Vertex Pharmaceuticals, Cambridge, MA, USA) is a small-molecule, selective, noncompetitive inhibitor directed against human IMPDH. In this report, we show that VX-944 inhibits in vitro growth of human multiple myeloma (MM) cell lines via induction of apoptosis. Interleukin-6, insulin-like growth factor-1, or co-culture with bone marrow stromal cells (BMSCs) do not protect against VX-944-induced MM cell growth inhibition. VX-944 induced apoptosis in MM cell lines with only modest activation of caspases 3, 8, and 9. Furthermore, the pan-caspase inhibitor z-VAD-fmk did not inhibit VX-944-induced apoptosis and cell death. During VX-944-induced apoptosis, expressions of Bax and Bak were enhanced, and both apoptosis-inducing factor (AIF) and endonuclease G (Endo G) were released from the mitochondria to cytosol, suggesting that VX-944 triggers apoptosis in MM cells primarily via a caspase-independent, Bax/AIF/Endo G pathway. Importantly, VX-944 augments the cytotoxicity of doxorubicin and melphalan even in the presence of BMSCs. Taken together, our data demonstrate a primarily non-caspase-dependent apoptotic pathway triggered by VX-944, thereby providing a rationale to enhance MM cell cytotoxicity by combining this agent with conventional agents which trigger caspase activation.
Collapse
Affiliation(s)
- Kenji Ishitsuka
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Moosavi MA, Yazdanparast R, Sanati MH. The Cytotoxic and Anti-proliferative Effects of 3-Hydrogenkwadaphnin in K562 and Jurkat Cells Is Reduced by Guanosine. BMB Rep 2005; 38:391-8. [PMID: 16053705 DOI: 10.5483/bmbrep.2005.38.4.391] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
3-hydrogenwadaphnin (3-HK) is a new daphnane-type diterpene ester isolated from Dendrostellera lessertii with strong anti-tumoral activity in animal models and in cultures. Here, prolonged effects of this new agent on proliferation and viability of several different cancerous cell lines were evaluated. Using [(3)H]thymidine incorporation, it was found that the drug inhibited cell proliferation and induced G1/S cell cycle arrest in leukemic cells 24 h after a single dose treatment. The cell viability of Jurkat cells was also decreased by almost 10 %, 31 % and 40 % after a single dose treatment (7.5 nM) at 24, 48 and 72 h, respectively. The drug-treated cells were stained with acridine orange/ ethidium bromide to document the chromatin condensation and DNA fragmentation. These observations were further confirmed by detection of DNA laddering pattern in the agarose gel electrophoresis of the extracted DNA from the treated cells. Treatment of K562 cells with the drug at 7.5, 15 and 30 nM caused apoptosis in 25 %, 45 % and 65 % of the cells, respectively. Exogenous addition of 25-50 microM guanosine and/or deoxyguanosine to the cell culture of the drug-treated cells restored DNA synthesis, released cell arrest at G1/S checkpoint and decreased the apoptotic cell death caused by the drug. These observations were not made using adenosine. However, the drug effects on K562 cells were potentiated by hypoxanthine. Based on these observations, perturbation of GTP metabolism is considered as one of the main reasons for apoptotic cell death by 3-HK.
Collapse
Affiliation(s)
- Mohammad Amin Moosavi
- Institute of Biochemistry and Biophysics, P. O. Box. 13145-1384, University of Tehran, Tehran, Iran
| | | | | |
Collapse
|
34
|
Gu JJ, Santiago L, Mitchell BS. Synergy between imatinib and mycophenolic acid in inducing apoptosis in cell lines expressing Bcr-Abl. Blood 2004; 105:3270-7. [PMID: 15604220 DOI: 10.1182/blood-2004-10-3864] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bcr-Abl tyrosine kinase activity initiates a number of intracellular signaling cascades that result in leukemogenesis. Imatinib mesylate, a specific Bcr-Abl tyrosine kinase inhibitor, has been highly successful in the treatment of chronic myelogenous leukemia (CML). However, the emergence of imatinib resistance and the incomplete molecular response of a significant number of patients receiving this therapy have led to a search for combinations of drugs that will enhance the efficacy of imatinib. We have demonstrated that mycophenolic acid (MPA), a specific inosine monophosphate dehydrogenase (IMPDH) inhibitor that results in depletion of intracellular guanine nucleotides, is synergistic with imatinib in inducing apoptosis in Bcr-Abl-expressing cell lines. Studies of signaling pathways downstream of Bcr-Abl demonstrated that the addition of MPA to imatinib reduced the phosphorylation of both Stat5 and Lyn, a Src kinase family member. The phosphorylation of S6 ribosomal protein was also greatly reduced. These results demonstrate that inhibitors of guanine nucleotide biosynthesis may synergize with imatinib in reducing the levels of minimal residual disease in CML and lay the foundation for clinical trials in which IMPDH inhibitors are added to imatinib in patients who have suboptimal molecular responses to single agent therapy or who have progressive disease.
Collapse
Affiliation(s)
- Jing Jin Gu
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599-7295, USA
| | | | | |
Collapse
|
35
|
Shipkova M, Spielbauer B, Voland A, Gröne HJ, Armstrong VW, Oellerich M, Wieland E. cDNA Microarray Analysis Reveals New Candidate Genes Possibly Linked to Side Effects Under Mycophenolate Mofetil Therapy. Transplantation 2004; 78:1145-52. [PMID: 15502711 DOI: 10.1097/01.tp.0000137262.06252.05] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Mycophenolate mofetil (MMF) the prodrug of mycophenolic acid is usually well tolerated. Side effects such as anemia and diarrhea occur in approximately 10%-15% of patients. The aim of this study was to examine in a rat model the effect of MMF on gene expression in liver and gut to identify target genes with possible relevance to MMF side effects. METHODS Twelve Wistar rats were treated with 40 mg/kg body weight MMF orally for 21 days. Controls (n=9) received vehicle only. RNA was extracted from liver, jejunum, ileum, and colon and transcribed into cDNA. Regulated genes were identified in liver by DNA microarray experiments. Gene regulation was verified in liver and gut using quantitative real-time PCR on the LightCycler instrument. Transcription elongation factor 2 served as reference gene. RESULTS Microarray analysis revealed that major alpha-hemoglobin, polymeric immunoglobulin receptor, catalase, and CCAAT/enhancer protein alpha gene expression were down-regulated in livers of MMF-treated rats 10-, 5.5-, 4-, and 5-fold, respectively. These findings could be confirmed through quantitative real-time PCR analysis of gene expression in liver, ileum, jejunum, and colon. CONCLUSION Using microarray analysis and a rat model four candidate genes which may be functionally linked to side effects (major alpha-hemoglobin-->anaemia; polymeric immunoglobulin receptor-->protection of mucosa; catalase and CCAAT/enhancer protein alpha-->oxidative stress) of MMF therapy were identified.
Collapse
Affiliation(s)
- Maria Shipkova
- Zentralinstitut für Klinische Chemie und Laboratoriumsmedizin, Klinikum Stuttgart, Katharinenhospital, Kriegsbergstrasse 60, D-70174 Stuttgart, Germany.
| | | | | | | | | | | | | |
Collapse
|