101
|
Sperling AS, Guerra VA, Kennedy JA, Yan Y, Hsu JI, Wang F, Nguyen AT, Miller PG, McConkey ME, Quevedo Barrios VA, Furudate K, Zhang L, Kanagal-Shamanna R, Zhang J, Little L, Gumbs C, Daver N, DiNardo CD, Kadia T, Ravandi F, Kantarjian H, Garcia-Manero G, Futreal PA, Ebert BL, Takahashi K. Lenalidomide promotes the development of TP53-mutated therapy-related myeloid neoplasms. Blood 2022; 140:1753-1763. [PMID: 35512188 PMCID: PMC9837415 DOI: 10.1182/blood.2021014956] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/25/2022] [Indexed: 01/26/2023] Open
Abstract
There is a growing body of evidence that therapy-related myeloid neoplasms (t-MNs) with driver gene mutations arise in the background of clonal hematopoiesis (CH) under the positive selective pressure of chemo- and radiation therapies. Uncovering the exposure relationships that provide selective advantage to specific CH mutations is critical to understanding the pathogenesis and etiology of t-MNs. In a systematic analysis of 416 patients with t-MN and detailed prior exposure history, we found that TP53 mutations were significantly associated with prior treatment with thalidomide analogs, specifically lenalidomide. We demonstrated experimentally that lenalidomide treatment provides a selective advantage to Trp53-mutant hematopoietic stem and progenitor cells (HSPCs) in vitro and in vivo, the effect of which was specific to Trp53-mutant HSPCs and was not observed in HSPCs with other CH mutations. Because of the differences in CK1α degradation, pomalidomide treatment did not provide an equivalent level of selective advantage to Trp53-mutant HSPCs, providing a biological rationale for its use in patients at high risk for t-MN. These findings highlight the role of lenalidomide treatment in promoting TP53-mutated t-MNs and offer a potential alternative strategy to mitigate the risk of t-MN development.
Collapse
Affiliation(s)
- Adam S. Sperling
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology, Brigham and Women’s Hospital, Boston, MA
| | - Veronica A. Guerra
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - James A. Kennedy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, University of Toronto, Toronto, Canada
- Division of Hematology and Medical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Division of Hematology and Medical Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Yuanqing Yan
- Department of Neurosurgery, University of Northwestern, Chicago, IL
| | - Joanne I. Hsu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Feng Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Andrew T. Nguyen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Peter G. Miller
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Marie E. McConkey
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Ken Furudate
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Oral and Maxillofacial Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Linda Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Latasha Little
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Curtis Gumbs
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Courtney D. DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Tapan Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - P. Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Benjamin L. Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Howard Hughes Medical Institute, Dana-Farber Cancer Institute, Boston, MA
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
102
|
Sainas S, Giorgis M, Circosta P, Poli G, Alberti M, Passoni A, Gaidano V, Pippione AC, Vitale N, Bonanni D, Rolando B, Cignetti A, Ramondetti C, Lanno A, Ferraris DM, Canepa B, Buccinnà B, Piccinini M, Rizzi M, Saglio G, Al-Karadaghi S, Boschi D, Miggiano R, Tuccinardi T, Lolli ML. Targeting Acute Myelogenous Leukemia Using Potent Human Dihydroorotate Dehydrogenase Inhibitors Based on the 2-Hydroxypyrazolo[1,5- a]pyridine Scaffold: SAR of the Aryloxyaryl Moiety. J Med Chem 2022; 65:12701-12724. [PMID: 36162075 PMCID: PMC9574863 DOI: 10.1021/acs.jmedchem.2c00496] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Indexed: 11/28/2022]
Abstract
In recent years, human dihydroorotate dehydrogenase inhibitors have been associated with acute myelogenous leukemia as well as studied as potent host targeting antivirals. Starting from MEDS433 (IC50 1.2 nM), we kept improving the structure-activity relationship of this class of compounds characterized by 2-hydroxypyrazolo[1,5-a]pyridine scaffold. Using an in silico/crystallography supported design, we identified compound 4 (IC50 7.2 nM), characterized by the presence of a decorated aryloxyaryl moiety that replaced the biphenyl scaffold, with potent inhibition and pro-differentiating abilities on AML THP1 cells (EC50 74 nM), superior to those of brequinar (EC50 249 nM) and boosted when in combination with dipyridamole. Finally, compound 4 has an extremely low cytotoxicity on non-AML cells as well as MEDS433; it has shown a significant antileukemic activity in vivo in a xenograft mouse model of AML.
Collapse
Affiliation(s)
- Stefano Sainas
- Department
of Drug Science and Technology, University
of Turin, Via P. Giuria 9, Turin 10125, Italy
| | - Marta Giorgis
- Department
of Drug Science and Technology, University
of Turin, Via P. Giuria 9, Turin 10125, Italy
| | - Paola Circosta
- Department
of Clinical and Biological Sciences, University
of Turin, Regione Gonzole 10, Orbassano, Turin 10043, Italy
- Molecular
Biotechnology Center, University of Turin, Via Nizza 52, Turin 10126, Italy
| | - Giulio Poli
- Department
of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Marta Alberti
- Department
of Pharmaceutical Sciences, University of
Piemonte Orientale, Via
G. Bovio 6, Novara 28100, Italy
| | - Alice Passoni
- Laboratory
of Mass Spectrometry, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan 20156, Italy
| | - Valentina Gaidano
- Division
of Hematology and Cell Therapy, AO Ordine
Mauriziano, Largo Filippo
Turati, 62, Turin 10128, Italy
| | - Agnese C. Pippione
- Department
of Drug Science and Technology, University
of Turin, Via P. Giuria 9, Turin 10125, Italy
| | - Nicoletta Vitale
- Department
of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, Turin 10126, Italy
| | - Davide Bonanni
- Department
of Drug Science and Technology, University
of Turin, Via P. Giuria 9, Turin 10125, Italy
- Life
Science Department, University of Modena, Via Università 4, Modena 41121, Italy
| | - Barbara Rolando
- Department
of Drug Science and Technology, University
of Turin, Via P. Giuria 9, Turin 10125, Italy
| | - Alessandro Cignetti
- Division
of Hematology and Cell Therapy, AO Ordine
Mauriziano, Largo Filippo
Turati, 62, Turin 10128, Italy
| | - Cristina Ramondetti
- Department
of Oncology, University of Turin, Via Michelangelo 27/B, Turin 10125, Italy
| | - Alessia Lanno
- Laboratory
of Mass Spectrometry, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan 20156, Italy
| | - Davide M. Ferraris
- Department
of Pharmaceutical Sciences, University of
Piemonte Orientale, Via
G. Bovio 6, Novara 28100, Italy
| | - Barbara Canepa
- GEM FORLAB, Via Ing.
Comotto 36, Caluso, Turin, 10014, Italy
| | - Barbara Buccinnà
- Department
of Oncology, University of Turin, Via Michelangelo 27/B, Turin 10125, Italy
| | - Marco Piccinini
- Department
of Oncology, University of Turin, Via Michelangelo 27/B, Turin 10125, Italy
| | - Menico Rizzi
- Department
of Pharmaceutical Sciences, University of
Piemonte Orientale, Via
G. Bovio 6, Novara 28100, Italy
| | - Giuseppe Saglio
- Department
of Clinical and Biological Sciences, University
of Turin, Regione Gonzole 10, Orbassano, Turin 10043, Italy
- Division
of Hematology and Cell Therapy, AO Ordine
Mauriziano, Largo Filippo
Turati, 62, Turin 10128, Italy
| | - Salam Al-Karadaghi
- Department
of Biochemistry and Structural Biology, Lund University, Naturvetarvägen
14, Box 124, Lund 221 00, Sweden
| | - Donatella Boschi
- Department
of Drug Science and Technology, University
of Turin, Via P. Giuria 9, Turin 10125, Italy
| | - Riccardo Miggiano
- Department
of Pharmaceutical Sciences, University of
Piemonte Orientale, Via
G. Bovio 6, Novara 28100, Italy
| | - Tiziano Tuccinardi
- Molecular
Biotechnology Center, University of Turin, Via Nizza 52, Turin 10126, Italy
| | - Marco L. Lolli
- Department
of Drug Science and Technology, University
of Turin, Via P. Giuria 9, Turin 10125, Italy
| |
Collapse
|
103
|
Darekar S, Laín S. Asymmetric inheritance of cytoophidia could contribute to determine cell fate and plasticity: The onset of alternative differentiation patterns in daughter cells may rely on the acquisition of either CTPS or IMPDH cytoophidia: The onset of alternative differentiation patterns in daughter cells may rely on the acquisition of either CTPS or IMPDH cytoophidia. Bioessays 2022; 44:e2200128. [PMID: 36209393 DOI: 10.1002/bies.202200128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/26/2022] [Accepted: 09/21/2022] [Indexed: 12/20/2022]
Abstract
Two enzymes involved in the synthesis of pyrimidine and purine nucleotides, CTP synthase (CTPS) and IMP dehydrogenase (IMPDH), can assemble into a single or very few large filaments called rods and rings (RR) or cytoophidia. Most recently, asymmetric cytoplasmic distribution of organelles during cell division has been described as a decisive event in hematopoietic stem cell fate. We propose that cytoophidia, which could be considered as membrane-less organelles, may also be distributed asymmetrically during mammalian cell division as previously described for Schizosaccharomyces pombe. Furthermore, because each type of nucleotide intervenes in distinct processes (e.g., membrane synthesis, glycosylation, and G protein-signaling), alterations in the rate of synthesis of specific nucleotide types could influence cell differentiation in multiple ways. Therefore, we hypothesize that whether a daughter cell inherits or not CTPS or IMPDH filaments determines its fate and that this asymmetric inheritance, together with the dynamic nature of these structures enables plasticity in a cell population.
Collapse
Affiliation(s)
- Suhas Darekar
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, Stockholm, Sweden
| | - Sonia Laín
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
104
|
Wang W, Li H, Huang M, Wang X, Li W, Qian X, Jing L. Hoxa9/ meis1-transgenic zebrafish develops acute myeloid leukaemia-like disease with rapid onset and high penetrance. Open Biol 2022; 12:220172. [PMID: 36285442 PMCID: PMC9597180 DOI: 10.1098/rsob.220172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022] Open
Abstract
HOXA9 and MEIS1 are co-expressed in over 50% of acute myeloid leukaemia (AML) and play essential roles in leukaemogenesis, but the mechanisms involved are poorly understood. Diverse animal models offer valuable tools to recapitulate different aspects of AML and link in vitro studies to clinical trials. We generated a double transgenic zebrafish that enables hoxa9 overexpression in blood cells under the draculin (drl) regulatory element and an inducible expression of meis1 through a heat shock promoter. After induction, Tg(drl:hoxa9;hsp70:meis1) embryos developed a preleukaemic state with reduced myeloid and erythroid differentiation coupled with the poor production of haematopoietic stem cells and myeloid progenitors. Importantly, most adult Tg(drl:hoxa9;hsp70:meis1) fish at 3 months old showed abundant accumulations of immature myeloid precursors, interrupted differentiation and anaemia in the kidney marrow, and infiltration of myeloid precursors in peripheral blood, resembling human AML. Genome-wide transcriptional analysis also confirmed AML transformation by the transgene. Moreover, the dihydroorotate dehydrogenase (DHODH) inhibitor that reduces leukaemogenesis in mammals effectively restored haematopoiesis in Tg(drl:hoxa9;hsp70:meis1) embryos and improved their late survival. Thus, Tg(drl:hoxa9;hsp70:meis1) zebrafish is a rapid-onset high-penetrance AML-like disease model, which provides a novel tool to harness the unique advantages of zebrafish for mechanistic studies and drug screening against HOXA9/MEIS1 overexpressed high-risk AML.
Collapse
Affiliation(s)
- Wei Wang
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Hongji Li
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Mengling Huang
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Xue Wang
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Wei Li
- Core facility and technical service center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Xiaoqing Qian
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Lili Jing
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| |
Collapse
|
105
|
Zhang P, Brinton LT, Gharghabi M, Sher S, Williams K, Cannon M, Walker JS, Canfield D, Beaver L, Cempre CB, Phillips H, Chen X, Yan P, Lehman A, Scherle P, Wang M, Vaddi K, Baiocchi R, Wang R, Sampath D, Alinari L, Blachly JS, Lapalombella R. Targeting OXPHOS de novo purine synthesis as the nexus of FLT3 inhibitor-mediated synergistic antileukemic actions. SCIENCE ADVANCES 2022; 8:eabp9005. [PMID: 36112677 PMCID: PMC9481139 DOI: 10.1126/sciadv.abp9005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 07/28/2022] [Indexed: 05/09/2023]
Abstract
Using a genome-wide CRISPR screen, we identified CDK9, DHODH, and PRMT5 as synthetic lethal partners with gilteritinib treatment in fms-like tyrosine kinase 3 (FLT3)-internal tandem duplication (ITD) acute myeloid leukemia (AML) and genetically and pharmacologically validated their roles in gilteritinib sensitivity. The presence of FLT3-ITD is associated with an increase in anaerobic glycolysis, rendering leukemia cells highly sensitive to inhibition of glycolysis. Supportive of this, our data show the enrichment of single guide RNAs targeting 28 glycolysis-related genes upon gilteritinib treatment, suggesting that switching from glycolysis to oxidative phosphorylation (OXPHOS) may represent a metabolic adaption of AML in gilteritinib resistance. CDK9i/FLT3i, DHODHi/FLT3i, and PRMT5i/FLT3i pairs mechanistically converge on OXPHOS and purine biosynthesis blockade, implying that targeting the metabolic functions of these three genes and/or proteins may represent attractive strategies to sensitize AML to gilteritinib treatment. Our findings provide the basis for maximizing therapeutic impact of FLT3-ITD inhibitors and a rationale for a clinical trial of these novel combinations.
Collapse
Affiliation(s)
- Pu Zhang
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Lindsey T. Brinton
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Mehdi Gharghabi
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
- Department of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Steven Sher
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Katie Williams
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Matthew Cannon
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Janek S. Walker
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Daniel Canfield
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Larry Beaver
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Casey B. Cempre
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Hannah Phillips
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Xuyong Chen
- Center for Childhood Cancer and Blood Diseases, Hematology/Oncology and BMT, Abigail Wexner Research Institute at Nationwide Children’s Hospital, The Ohio State University, Columbus, OH, USA
| | - Pearlly Yan
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Amy Lehman
- Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | | | - Min Wang
- Prelude Therapeutics, Wilmington, DE, USA
| | - Kris Vaddi
- Prelude Therapeutics, Wilmington, DE, USA
| | - Robert Baiocchi
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Ruoning Wang
- Center for Childhood Cancer and Blood Diseases, Hematology/Oncology and BMT, Abigail Wexner Research Institute at Nationwide Children’s Hospital, The Ohio State University, Columbus, OH, USA
| | - Deepa Sampath
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Lapo Alinari
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - James S. Blachly
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
- Leukemia Research Program, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| | - Rosa Lapalombella
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
- Leukemia Research Program, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
106
|
Li C, Zhou Y, Xu J, Zhou X, Liu S, Huang Z, Qiu Z, Zeng T, Gou K, Tao L, Zhong X, Yang X, Zhou Y, Su N, Chen Q, Zhao Y, Luo Y. Discovery of potent human dihydroorotate dehydrogenase inhibitors based on a benzophenone scaffold. Eur J Med Chem 2022; 243:114737. [PMID: 36115209 DOI: 10.1016/j.ejmech.2022.114737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/20/2022] [Accepted: 08/29/2022] [Indexed: 12/26/2022]
Abstract
Blocking the de novo biosynthesis of pyrimidine by inhibiting human dihydroorotate dehydrogenase (hDHODH) is an effective way to suppress the proliferation of cancer cells and activated lymphocytes. Herein, eighteen teriflunomide derivatives and four ASLAN003 derivatives were designed and synthesized as novel hDHODH inhibitors based on a benzophenone scaffold. The optimal compound 7d showed a potent hDHODH inhibitory activity with an IC50 value of 10.9 nM, and displayed promising antiproliferative activities against multiple human cancer cells with IC50 values of 0.1-0.8 μM. Supplementation of exogenous uridine rescued the cell viability of 7d-treated Raji and HCT116 cells. Meanwhile, 7d significantly induced cell cycle S-phase arrest in Raji and HCT116 cells. Furthermore, 7d exhibited favorable safety profiles in mice and displayed effective antitumor activities with tumor growth inhibition (TGI) rates of 58.3% and 42.1% at an oral dosage of 30 mg/kg in Raji and HCT116 cells xenograft models, respectively. Taken together, these findings provide a promising hDHODH inhibitor 7d with potential activities against some tumors.
Collapse
Affiliation(s)
- Chungen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Yue Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Jing Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Xia Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Song Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Zongkai Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Zhiqiang Qiu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Ting Zeng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Kun Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Lei Tao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Xi Zhong
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Xiaowei Yang
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yang Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Na Su
- Department of Pharmacy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Qiang Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Yinglan Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China; Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - Youfu Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
107
|
Demarest JF, Kienle M, Boytz R, Ayres M, Kim EJ, Patten JJ, Chung D, Gandhi V, Davey R, Sykes DB, Shohdy N, Pottage JC, Kumar VS. Brequinar and dipyridamole in combination exhibits synergistic antiviral activity against SARS-CoV-2 in vitro: Rationale for a host-acting antiviral treatment strategy for COVID-19. Antiviral Res 2022; 206:105403. [PMID: 36041646 PMCID: PMC9420051 DOI: 10.1016/j.antiviral.2022.105403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/28/2022] [Accepted: 08/23/2022] [Indexed: 11/25/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19) and the associated global pandemic resulting in >400 million infections worldwide and several million deaths. The continued evolution of SARS-CoV-2 to potentially evade vaccines and monoclonal antibody (mAb)-based therapies and the limited number of authorized small-molecule antivirals necessitates the need for development of new drug treatments. There remains an unmet medical need for effective and convenient treatment options for SARS-CoV-2 infection. SARS-CoV-2 is an RNA virus that depends on host intracellular ribonucleotide pools for its replication. Dihydroorotate dehydrogenase (DHODH) is a ubiquitous host enzyme that is required for de novo pyrimidine synthesis. The inhibition of DHODH leads to a depletion of intracellular pyrimidines, thereby impacting viral replication in vitro. Brequinar (BRQ) is an orally available, selective, and potent low nanomolar inhibitor of human DHODH that has been shown to exhibit broad spectrum inhibition of RNA virus replication. However, host cell nucleotide salvage pathways can maintain intracellular pyrimidine levels and compensate for BRQ-mediated DHODH inhibition. In this report, we show that the combination of BRQ and the salvage pathway inhibitor dipyridamole (DPY) exhibits strong synergistic antiviral activity in vitro against SARS-CoV-2 by enhanced depletion of the cellular pyrimidine nucleotide pool. The combination of BRQ and DPY showed antiviral activity against the prototype SARS-CoV-2 as well as the Beta (B.1.351) and Delta (B.1.617.2) variants. These data support the continued evaluation of the combination of BRQ and DPY as a broad-spectrum, host-acting antiviral strategy to treat SARS-CoV-2 and potentially other RNA virus infections.
Collapse
Affiliation(s)
| | | | | | - Mary Ayres
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | - Varsha Gandhi
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | |
Collapse
|
108
|
Jackson TR, Vuorinen A, Josa-Culleré L, Madden KS, Conole D, Cogswell TJ, Wilkinson IV, Kettyle LM, Zhang D, O’Mahony A, Gracias D, McCall L, Westwood R, Terstappen GC, Davies SG, Tate EW, Wynne GM, Vyas P, Russell AJ, Milne TA. A tubulin binding molecule drives differentiation of acute myeloid leukemia cells. iScience 2022; 25:104787. [PMID: 35992086 PMCID: PMC9385704 DOI: 10.1016/j.isci.2022.104787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/31/2022] [Accepted: 07/13/2022] [Indexed: 12/01/2022] Open
Abstract
Despite much progress in developing better drugs, many patients with acute myeloid leukemia (AML) still die within a year of diagnosis. This is partly because it is difficult to identify therapeutic targets that are effective across multiple AML subtypes. One common factor across AML subtypes is the presence of a block in differentiation. Overcoming this block should allow for the identification of therapies that are not dependent on a specific mutation for their efficacy. Here, we used a phenotypic screen to identify compounds that stimulate differentiation in genetically diverse AML cell lines. Lead compounds were shown to decrease tumor burden and to increase survival in vivo. Using multiple complementary target deconvolution approaches, these compounds were revealed to be anti-mitotic tubulin disruptors that cause differentiation by inducing a G2-M mitotic arrest. Together, these results reveal a function for tubulin disruptors in causing differentiation of AML cells.
Collapse
Affiliation(s)
- Thomas R. Jackson
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Aini Vuorinen
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Laia Josa-Culleré
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - Katrina S. Madden
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - Daniel Conole
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, 82 Wood Lane, London W12 0BZ, UK
| | - Thomas J. Cogswell
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Isabel V.L. Wilkinson
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - Laura M. Kettyle
- Axis Bioservices, 189 Castleroe Rd, Coleraine, Co. Londonderry BT51 3RP, Northern Ireland
| | - Douzi Zhang
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Alison O’Mahony
- Eurofins Discovery Phenotypic Services, St. Charles, MO 63304 and Burlingame, CA 94010, USA
- Discovery Platform at Recursion, 41 S Rio Grande Street, Salt Lake City, UT 84101, USA
| | - Deanne Gracias
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Lorna McCall
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Robert Westwood
- Oxstem Ltd, Midland House West Way, Botley, Oxford OX2 0PH, UK
| | | | - Stephen G. Davies
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - Edward W. Tate
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, 82 Wood Lane, London W12 0BZ, UK
| | - Graham M. Wynne
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
- Oxstem Ltd, Midland House West Way, Botley, Oxford OX2 0PH, UK
| | - Paresh Vyas
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Angela J. Russell
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Thomas A. Milne
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
109
|
Olsen TK, Dyberg C, Embaie BT, Alchahin A, Milosevic J, Ding J, Otte J, Tümmler C, Hed Myrberg I, Westerhout EM, Koster J, Versteeg R, Ding HF, Kogner P, Johnsen JI, Sykes DB, Baryawno N. DHODH is an independent prognostic marker and potent therapeutic target in neuroblastoma. JCI Insight 2022; 7:153836. [PMID: 35943801 PMCID: PMC9798925 DOI: 10.1172/jci.insight.153836] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 08/04/2022] [Indexed: 01/11/2023] Open
Abstract
Despite intensive therapy, children with high-risk neuroblastoma are at risk of treatment failure. We applied a multiomic system approach to evaluate metabolic vulnerabilities in human neuroblastoma. We combined metabolomics, CRISPR screening, and transcriptomic data across more than 700 solid tumor cell lines and identified dihydroorotate dehydrogenase (DHODH), a critical enzyme in pyrimidine synthesis, as a potential treatment target. Of note, DHODH inhibition is currently under clinical investigation in patients with hematologic malignancies. In neuroblastoma, DHODH expression was identified as an independent risk factor for aggressive disease, and high DHODH levels correlated to worse overall and event-free survival. A subset of tumors with the highest DHODH expression was associated with a dismal prognosis, with a 5-year survival of less than 10%. In xenograft and transgenic neuroblastoma mouse models treated with the DHODH inhibitor brequinar, tumor growth was dramatically reduced, and survival was extended. Furthermore, brequinar treatment was shown to reduce the expression of MYC targets in 3 neuroblastoma models in vivo. A combination of brequinar and temozolomide was curative in the majority of transgenic TH-MYCN neuroblastoma mice, indicating a highly active clinical combination therapy. Overall, DHODH inhibition combined with temozolomide has therapeutic potential in neuroblastoma, and we propose this combination for clinical testing.
Collapse
Affiliation(s)
- Thale Kristin Olsen
- Division of Pediatric Oncology and Pediatric Surgery, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden.,Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Cecilia Dyberg
- Division of Pediatric Oncology and Pediatric Surgery, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Bethel Tesfai Embaie
- Division of Pediatric Oncology and Pediatric Surgery, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Adele Alchahin
- Division of Pediatric Oncology and Pediatric Surgery, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Jelena Milosevic
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jane Ding
- Division of Molecular and Cellular Pathology, Department of Pathology, Heersink School of Medicine, the University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jörg Otte
- Division of Pediatric Oncology and Pediatric Surgery, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Conny Tümmler
- Division of Pediatric Oncology and Pediatric Surgery, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Ida Hed Myrberg
- Division of Pediatric Oncology and Pediatric Surgery, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Ellen M. Westerhout
- Department of Oncogenomics, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Jan Koster
- Department of Oncogenomics, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Rogier Versteeg
- Department of Oncogenomics, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Han-Fei Ding
- Division of Molecular and Cellular Pathology, Department of Pathology, Heersink School of Medicine, the University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Per Kogner
- Division of Pediatric Oncology and Pediatric Surgery, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - John Inge Johnsen
- Division of Pediatric Oncology and Pediatric Surgery, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - David B. Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Ninib Baryawno
- Division of Pediatric Oncology and Pediatric Surgery, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
110
|
Cisar JS, Pietsch C, DeRatt LG, Jacoby E, Kazmi F, Keohane C, Legenski K, Matico R, Shaffer P, Simonnet Y, Tanner A, Wang CY, Wang W, Attar R, Edwards JP, Kuduk SD. N-Heterocyclic 3-Pyridyl Carboxamide Inhibitors of DHODH for the Treatment of Acute Myelogenous Leukemia. J Med Chem 2022; 65:11241-11256. [PMID: 35925768 DOI: 10.1021/acs.jmedchem.2c00788] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Acute myelogenous leukemia (AML), a disease of the blood and bone marrow, is characterized by the inability of myeloblasts to differentiate into mature cell types. Dihydroorotate dehydrogenase (DHODH) is an enzyme well-known in the pyrimidine biosynthesis pathway; however, small molecule DHODH inhibitors were recently shown to induce differentiation in multiple AML subtypes. Using virtual screening and structure-based drug design approaches, a new series of N-heterocyclic 3-pyridyl carboxamide DHODH inhibitors were discovered. Two lead compounds, 19 and 29, have potent biochemical and cellular DHODH activity, favorable physicochemical properties, and efficacy in a preclinical model of AML.
Collapse
Affiliation(s)
- Justin S Cisar
- Janssen Research and Development, 1400 McKean Rd, Spring House, Pennsylvania 19477, United States
| | - Christine Pietsch
- Janssen Research and Development, 1400 McKean Rd, Spring House, Pennsylvania 19477, United States
| | - Lindsey G DeRatt
- Janssen Research and Development, 1400 McKean Rd, Spring House, Pennsylvania 19477, United States
| | - Edgar Jacoby
- Janssen Research and Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Faraz Kazmi
- Janssen Research and Development, 1400 McKean Rd, Spring House, Pennsylvania 19477, United States
| | - Colleen Keohane
- Janssen Research and Development, 1400 McKean Rd, Spring House, Pennsylvania 19477, United States
| | - Katie Legenski
- Janssen Research and Development, 1400 McKean Rd, Spring House, Pennsylvania 19477, United States
| | - Rosalie Matico
- Janssen Research and Development, 1400 McKean Rd, Spring House, Pennsylvania 19477, United States
| | - Paul Shaffer
- Janssen Research and Development, 1400 McKean Rd, Spring House, Pennsylvania 19477, United States
| | - Yvan Simonnet
- Janssen Research and Development, 1400 McKean Rd, Spring House, Pennsylvania 19477, United States
| | - Alexandra Tanner
- Janssen Research and Development, 1400 McKean Rd, Spring House, Pennsylvania 19477, United States
| | - Chao-Yuan Wang
- Janssen Research and Development, 1400 McKean Rd, Spring House, Pennsylvania 19477, United States
| | - Weixue Wang
- Janssen Research and Development, 1400 McKean Rd, Spring House, Pennsylvania 19477, United States
| | - Ricardo Attar
- Janssen Research and Development, 1400 McKean Rd, Spring House, Pennsylvania 19477, United States
| | - James P Edwards
- Janssen Research and Development, San Diego, California 92121, United States
| | - Scott D Kuduk
- Janssen Research and Development, 1400 McKean Rd, Spring House, Pennsylvania 19477, United States
| |
Collapse
|
111
|
Diehl FF, Miettinen TP, Elbashir R, Nabel CS, Darnell AM, Do BT, Manalis SR, Lewis CA, Vander Heiden MG. Nucleotide imbalance decouples cell growth from cell proliferation. Nat Cell Biol 2022; 24:1252-1264. [PMID: 35927450 PMCID: PMC9359916 DOI: 10.1038/s41556-022-00965-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 06/21/2022] [Indexed: 12/26/2022]
Abstract
Nucleotide metabolism supports RNA synthesis and DNA replication to enable cell growth and division. Nucleotide depletion can inhibit cell growth and proliferation, but how cells sense and respond to changes in the relative levels of individual nucleotides is unclear. Moreover, the nucleotide requirement for biomass production changes over the course of the cell cycle, and how cells coordinate differential nucleotide demands with cell cycle progression is not well understood. Here we find that excess levels of individual nucleotides can inhibit proliferation by disrupting the relative levels of nucleotide bases needed for DNA replication and impeding DNA replication. The resulting purine and pyrimidine imbalances are not sensed by canonical growth regulatory pathways like mTORC1, Akt and AMPK signalling cascades, causing excessive cell growth despite inhibited proliferation. Instead, cells rely on replication stress signalling to survive during, and recover from, nucleotide imbalance during S phase. We find that ATR-dependent replication stress signalling is activated during unperturbed S phases and promotes nucleotide availability to support DNA replication. Together, these data reveal that imbalanced nucleotide levels are not detected until S phase, rendering cells reliant on replication stress signalling to cope with this metabolic problem and disrupting the coordination of cell growth and division.
Collapse
Affiliation(s)
- Frances F Diehl
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Teemu P Miettinen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Ryan Elbashir
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christopher S Nabel
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Alicia M Darnell
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brian T Do
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Health Sciences and Technology, Cambridge, MA, USA
| | - Scott R Manalis
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Departments of Biological Engineering and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
112
|
Teriflunomide Loaded SPION Nanoparticles Induced Apoptosis in MDA-MB-231 Breast Cancer Cells. J CLUST SCI 2022. [DOI: 10.1007/s10876-022-02327-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
113
|
Abohassan M, Al Shahrani M, Rajagopalan P. Identification of second generation benzylidene chromanone analogues as novel, potent DHODH inhibitors in acute myeloid leukemia cells. J Biomol Struct Dyn 2022:1-10. [DOI: 10.1080/07391102.2022.2103031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mesfer Al Shahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Prasanna Rajagopalan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
- Central Research Laboratory, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
114
|
CHST15 gene germline mutation is associated with the development of familial myeloproliferative neoplasms and higher transformation risk. Cell Death Dis 2022; 13:586. [PMID: 35798703 PMCID: PMC9263130 DOI: 10.1038/s41419-022-05035-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/15/2022] [Accepted: 06/22/2022] [Indexed: 01/21/2023]
Abstract
Herein, we describe the clinical and hematological features of three genetically related families predisposed to myeloproliferative neoplasms (MPNs). Using whole-exome sequencing, we identified a c.1367delG mutation(p.Arg456fs) in CHST15 (NM_001270764), a gene encoding a type II transmembraneglycoproteinthat acts as a sulfotransferase and participates in the biosynthesis of chondroitin sulfate E, in germline and somatic cells in familial MPN. CHST15defects caused an increased JAK2V617F allele burden and upregulated p-Stat3 activity,leading to an increase in the proliferative and prodifferentiation potential of transgenic HEL cells. We demonstrated that mutant CHST15 is able to coimmmunoprecipitate the JAK2 protein,suggesting the presence of a CHST15-JAK2-Stat3 signaling axis in familial MPN. Gene expression profiling showed that the FREM1, IFI27 and C4B_2 genes are overexpressed in familial MPN, suggesting the activation of an "inflammatory response-extracellular matrix-immune regulation" signaling network in the CHST15 mutation background.We thus concluded that CHST15 is a novel gene that predisposes to familial MPN and increases the probability of disease development or transformation.
Collapse
|
115
|
In vivo genome-wide CRISPR screening in murine acute myeloid leukemia uncovers microenvironmental dependencies. Blood Adv 2022; 6:5072-5084. [PMID: 35793392 PMCID: PMC9631646 DOI: 10.1182/bloodadvances.2022007250] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/26/2022] [Indexed: 11/20/2022] Open
Abstract
In vivo CRISPR screens in AML define key interactors of the microenvironment, including integrins, immune modulators, and glycosylation. Eight in vivo–specific hits are recurrently associated with adverse prognosis: BTBD6, FERMT3, ILK, SLC19A1, TAP2, TLN1, TPST2, and TRMT12.
Genome-wide CRISPR screens have been extremely useful in identifying therapeutic targets in diverse cancers by defining genes that are essential for malignant growth. However, most CRISPR screens were performed in vitro and thus cannot identify genes that are essential for interactions with the microenvironment in vivo. Here, we report genome-wide CRISPR screens in 2 in vivo murine models of acute myeloid leukemia (AML) driven by the KMT2A/MLLT3 fusion or by the constitutive coexpression of Hoxa9 and Meis1. Secondary validation using a focused library identified 72 genes specifically essential for leukemic growth in vivo, including components of the major histocompatibility complex class I complex, Cd47, complement receptor Cr1l, and the β-4-galactosylation pathway. Importantly, several of these in vivo–specific hits have a prognostic effect or are inferred to be master regulators of protein activity in human AML cases. For instance, we identified Fermt3, a master regulator of integrin signaling, as having in vivo–specific dependency with high prognostic relevance. Overall, we show an experimental and computational pipeline for genome-wide functional screens in vivo in AML and provide a genome-wide resource of essential drivers of leukemic growth in vivo.
Collapse
|
116
|
Alqahtani SS, Koltai T, Ibrahim ME, Bashir AHH, Alhoufie STS, Ahmed SBM, Molfetta DD, Carvalho TMA, Cardone RA, Reshkin SJ, Hifny A, Ahmed ME, Alfarouk KO. Role of pH in Regulating Cancer Pyrimidine Synthesis. J Xenobiot 2022; 12:158-180. [PMID: 35893264 PMCID: PMC9326563 DOI: 10.3390/jox12030014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/17/2022] [Accepted: 06/28/2022] [Indexed: 11/18/2022] Open
Abstract
Replication is a fundamental aspect of cancer, and replication is about reproducing all the elements and structures that form a cell. Among them are DNA, RNA, enzymes, and coenzymes. All the DNA is doubled during each S (synthesis) cell cycle phase. This means that six billion nucleic acids must be synthesized in each cycle. Tumor growth, proliferation, and mutations all depend on this synthesis. Cancer cells require a constant supply of nucleotides and other macromolecules. For this reason, they must stimulate de novo nucleotide synthesis to support nucleic acid provision. When deregulated, de novo nucleic acid synthesis is controlled by oncogenes and tumor suppressor genes that enable increased synthesis and cell proliferation. Furthermore, cell duplication must be achieved swiftly (in a few hours) and in the midst of a nutrient-depleted and hypoxic environment. This also means that the enzymes participating in nucleic acid synthesis must work efficiently. pH is a critical factor in enzymatic efficiency and speed. This review will show that the enzymatic machinery working in nucleic acid synthesis requires a pH on the alkaline side in most cases. This coincides with many other pro-tumoral factors, such as the glycolytic phenotype, benefiting from an increased intracellular pH. An increased intracellular pH is a perfect milieu for high de novo nucleic acid production through optimal enzymatic performance.
Collapse
Affiliation(s)
- Saad Saeed Alqahtani
- Department of Pharmacy Practice, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
- Pharmacy Practice Research Unit, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | | | - Muntaser E. Ibrahim
- Department of Molecular Biology, Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan; (M.E.I.); (A.H.H.B.)
| | - Adil H. H. Bashir
- Department of Molecular Biology, Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan; (M.E.I.); (A.H.H.B.)
| | - Sari T. S. Alhoufie
- Medical Laboratories Technology Department, College of Applied Medical Sciences, Taibah University, Medina 42353, Saudi Arabia;
| | - Samrein B. M. Ahmed
- Department of Biosciences and Chemistry, College of Health, Wellbeing and Life Sciences, Sheffield Hallam University, Sheffield S1 1WB, UK;
| | - Daria Di Molfetta
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (T.M.A.C.); (R.A.C.); (S.J.R.)
| | - Tiago M. A. Carvalho
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (T.M.A.C.); (R.A.C.); (S.J.R.)
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (T.M.A.C.); (R.A.C.); (S.J.R.)
| | - Stephan Joel Reshkin
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (T.M.A.C.); (R.A.C.); (S.J.R.)
| | | | - Mohamed E. Ahmed
- Research Center, Zamzam University College, Khartoum 11123, Sudan;
| | - Khalid Omer Alfarouk
- Research Center, Zamzam University College, Khartoum 11123, Sudan;
- Alfarouk Biomedical Research LLC, Temple Terrace, FL 33617, USA
- Hala Alfarouk Cancer Center, Khartoum 11123, Sudan
- Correspondence:
| |
Collapse
|
117
|
Tomic B, Smoljo T, Lalic H, Dembitz V, Batinic J, Batinic D, Bedalov A, Visnjic D. Cytarabine-induced differentiation of AML cells depends on Chk1 activation and shares the mechanism with inhibitors of DHODH and pyrimidine synthesis. Sci Rep 2022; 12:11344. [PMID: 35790845 PMCID: PMC9256737 DOI: 10.1038/s41598-022-15520-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 06/24/2022] [Indexed: 01/19/2023] Open
Abstract
Acute myeloid leukemia (AML) is characterized by arrested differentiation making differentiation therapy a promising treatment strategy. Recent success of inhibitors of mutated isocitrate dehydrogenase (IDH) invigorated interest in differentiation therapy of AML so that several new drugs have been proposed, including inhibitors of dihydroorotate dehydrogenase (DHODH), an enzyme in pyrimidine synthesis. Cytarabine, a backbone of standard AML therapy, is known to induce differentiation at low doses, but the mechanism is not completely elucidated. We have previously reported that 5-aminoimidazole-4-carboxamide ribonucleoside (AICAr) and brequinar, a DHODH inhibitor, induced differentiation of myeloid leukemia by activating the ataxia telangiectasia and Rad3-related (ATR)/checkpoint kinase 1 (Chk1) via pyrimidine depletion. In this study, using immunoblotting, flow cytometry analyses, pharmacologic inhibitors and genetic inactivation of Chk1 in myeloid leukemia cell lines, we show that low dose cytarabine induces differentiation by activating Chk1. In addition, cytarabine induces differentiation ex vivo in a subset of primary AML samples that are sensitive to AICAr and DHODH inhibitor. The results of our study suggest that leukemic cell differentiation stimulated by low doses of cytarabine depends on the activation of Chk1 and thus shares the same pathway as pyrimidine synthesis inhibitors.
Collapse
Affiliation(s)
- Barbara Tomic
- grid.4808.40000 0001 0657 4636Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 3, 10 000 Zagreb, Croatia ,grid.4808.40000 0001 0657 4636Department of Physiology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Tomislav Smoljo
- grid.4808.40000 0001 0657 4636Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 3, 10 000 Zagreb, Croatia ,grid.4808.40000 0001 0657 4636Department of Physiology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Hrvoje Lalic
- grid.4808.40000 0001 0657 4636Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 3, 10 000 Zagreb, Croatia ,grid.4808.40000 0001 0657 4636Department of Physiology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Vilma Dembitz
- grid.4808.40000 0001 0657 4636Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 3, 10 000 Zagreb, Croatia ,grid.4808.40000 0001 0657 4636Department of Physiology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Josip Batinic
- grid.412688.10000 0004 0397 9648Division of Hematology, Department of Internal Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Drago Batinic
- grid.4808.40000 0001 0657 4636Department of Physiology, University of Zagreb School of Medicine, Zagreb, Croatia ,grid.412688.10000 0004 0397 9648Department of Laboratory Immunology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Antonio Bedalov
- grid.270240.30000 0001 2180 1622Clinical Research Division, Fred Hutchinson Cancer Research Centre, Seattle, WA USA
| | - Dora Visnjic
- grid.4808.40000 0001 0657 4636Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 3, 10 000 Zagreb, Croatia ,grid.4808.40000 0001 0657 4636Department of Physiology, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
118
|
Wang H, He X, Zhang L, Dong H, Huang F, Xian J, Li M, Chen W, Lu X, Pathak KV, Huang W, Li Z, Zhang L, Nguyen LXT, Yang L, Feng L, Gordon DJ, Zhang J, Pirrotte P, Chen CW, Salhotra A, Kuo YH, Horne D, Marcucci G, Sykes DB, Tiziani S, Jin H, Wang X, Li L. Disruption of dNTP homeostasis by ribonucleotide reductase hyperactivation overcomes AML differentiation blockade. Blood 2022; 139:3752-3770. [PMID: 35439288 PMCID: PMC9247363 DOI: 10.1182/blood.2021015108] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 04/07/2022] [Indexed: 01/09/2023] Open
Abstract
Differentiation blockade is a hallmark of acute myeloid leukemia (AML). A strategy to overcome such a blockade is a promising approach against the disease. The lack of understanding of the underlying mechanisms hampers development of such strategies. Dysregulated ribonucleotide reductase (RNR) is considered a druggable target in proliferative cancers susceptible to deoxynucleoside triphosphate (dNTP) depletion. Herein, we report an unanticipated discovery that hyperactivating RNR enables differentiation and decreases leukemia cell growth. We integrate pharmacogenomics and metabolomics analyses to identify that pharmacologically (eg, nelarabine) or genetically upregulating RNR subunit M2 (RRM2) creates a dNTP pool imbalance and overcomes differentiation arrest. Moreover, R-loop-mediated DNA replication stress signaling is responsible for RRM2 activation by nelarabine treatment. Further aggravating dNTP imbalance by depleting the dNTP hydrolase SAM domain and HD domain-containing protein 1 (SAMHD1) enhances ablation of leukemia stem cells by RRM2 hyperactivation. Mechanistically, excessive activation of extracellular signal-regulated kinase (ERK) signaling downstream of the imbalance contributes to cellular outcomes of RNR hyperactivation. A CRISPR screen identifies a synthetic lethal interaction between loss of DUSP6, an ERK-negative regulator, and nelarabine treatment. These data demonstrate that dNTP homeostasis governs leukemia maintenance, and a combination of DUSP inhibition and nelarabine represents a therapeutic strategy.
Collapse
Affiliation(s)
- Hanying Wang
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
- Department of Medical Oncology and
| | - Xin He
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Lei Zhang
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Haojie Dong
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Feiteng Huang
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Jie Xian
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Min Li
- Department of Information Sciences, Beckman Research Institute and
| | - Wei Chen
- Integrative Genomics Core, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Xiyuan Lu
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX
| | - Khyatiben V Pathak
- Cancer & Cell Biology Division, The Translational Genomics Research Institute, Phoenix, AZ
| | - Wenfeng Huang
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Zheng Li
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lianjun Zhang
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Le Xuan Truong Nguyen
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Lu Yang
- Department of Systems Biology, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Lifeng Feng
- Laboratory of Cancer Biology, Provincial Key Laboratory of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - David J Gordon
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Iowa, Iowa City, IA
| | - Jing Zhang
- McArdle Laboratory for Cancer Research and Wisconsin Blood Cancer Research Institute, University of Wisconsin-Madison, Madison, WI
| | - Patrick Pirrotte
- Cancer & Cell Biology Division, The Translational Genomics Research Institute, Phoenix, AZ
- Cancer & Cell Biology Division, The Translational Genomics Research Institute, Phoenix, AZ
| | - Chun-Wei Chen
- Department of Systems Biology, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | | | - Ya-Huei Kuo
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA
| | - Guido Marcucci
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
- Department of Hematology and Hematopoietic Cell Transplantation and
| | - David B Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA; and
| | - Stefano Tiziani
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX
- Department of Pediatrics and
- Department of Oncology, Dell Medical School, LiveSTRONG Cancer Institutes, The University of Texas at Austin, Austin, TX
| | - Hongchuan Jin
- Laboratory of Cancer Biology, Provincial Key Laboratory of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | | | - Ling Li
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| |
Collapse
|
119
|
Dihydroorotate dehydrogenase inhibition reveals metabolic vulnerability in chronic myeloid leukemia. Cell Death Dis 2022; 13:576. [PMID: 35773274 PMCID: PMC9247109 DOI: 10.1038/s41419-022-05028-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 06/12/2022] [Accepted: 06/15/2022] [Indexed: 01/21/2023]
Abstract
The development of different generations of BCR-ABL1 tyrosine kinase inhibitors (TKIs) has led to the high overall survival of chronic myeloid leukemia (CML) patients. However, there are CML patients who show resistance to TKI therapy and are prone to progress to more advanced phases of the disease. So, implementing an alternative approach for targeting TKIs insensitive cells would be of the essence. Dihydroorotate dehydrogenase (DHODH) is an enzyme in the de novo pyrimidine biosynthesis pathway that is located in the inner membrane of mitochondria. Here, we found that CML cells are vulnerable to DHODH inhibition mediated by Meds433, a new and potent DHODH inhibitor recently developed by our group. Meds433 significantly activates the apoptotic pathway and leads to the reduction of amino acids and induction of huge metabolic stress in CML CD34+ cells. Altogether, our study shows that DHODH inhibition is a promising approach for targeting CML stem/progenitor cells and may help more patients discontinue the therapy.
Collapse
|
120
|
Salame N, Fooks K, El-Hachem N, Bikorimana JP, Mercier FE, Rafei M. Recent Advances in Cancer Drug Discovery Through the Use of Phenotypic Reporter Systems, Connectivity Mapping, and Pooled CRISPR Screening. Front Pharmacol 2022; 13:852143. [PMID: 35795568 PMCID: PMC9250974 DOI: 10.3389/fphar.2022.852143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Multi-omic approaches offer an unprecedented overview of the development, plasticity, and resistance of cancer. However, the translation from anti-cancer compounds identified in vitro to clinically active drugs have a notoriously low success rate. Here, we review how technical advances in cell culture, robotics, computational biology, and development of reporter systems have transformed drug discovery, enabling screening approaches tailored to clinically relevant functional readouts (e.g., bypassing drug resistance). Illustrating with selected examples of “success stories,” we describe the process of phenotype-based high-throughput drug screening to target malignant cells or the immune system. Second, we describe computational approaches that link transcriptomic profiling of cancers with existing pharmaceutical compounds to accelerate drug repurposing. Finally, we review how CRISPR-based screening can be applied for the discovery of mechanisms of drug resistance and sensitization. Overall, we explore how the complementary strengths of each of these approaches allow them to transform the paradigm of pre-clinical drug development.
Collapse
Affiliation(s)
- Natasha Salame
- Department of Biomedical Sciences, Université de Montréal, Montreal, QC, Canada
| | - Katharine Fooks
- Lady Davis Institute for Medical Research, Montreal, QC, Canada
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - Nehme El-Hachem
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
| | - Jean-Pierre Bikorimana
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada
| | - François E. Mercier
- Lady Davis Institute for Medical Research, Montreal, QC, Canada
- Department of Medicine, McGill University, Montreal, QC, Canada
- *Correspondence: François E. Mercier, ; Moutih Rafei,
| | - Moutih Rafei
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada
- Molecular Biology Program, Université de Montréal, Montreal, QC, Canada
- *Correspondence: François E. Mercier, ; Moutih Rafei,
| |
Collapse
|
121
|
Matchett EC, Ambrose EC, Kornbluth J. Characterization of uridine-cytidine kinase like-1 nucleoside kinase activity and its role in tumor growth. Biochem J 2022; 479:1149-1164. [PMID: 35583288 PMCID: PMC9246348 DOI: 10.1042/bcj20210770] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 04/26/2022] [Accepted: 05/18/2022] [Indexed: 11/27/2022]
Abstract
Uridine-cytidine kinase like-1 (UCKL-1) is a largely uncharacterized protein with high sequence similarity to other uridine-cytidine kinases (UCKs). UCKs play an important role in the pyrimidine salvage pathway, catalyzing the phosphorylation of uridine and cytidine to UMP and CMP, respectively. Only two human UCKs have been identified, UCK1 and UCK2. Previous studies have shown both enzymes phosphorylate uridine and cytidine using ATP as the phosphate donor. No studies have evaluated the kinase potential of UCKL-1. We cloned and purified UCKL-1 and found that it successfully phosphorylated uridine and cytidine using ATP as the phosphate donor. The catalytic efficiency (calculated as kcat/KM) was 1.2 × 104 s-1, M-1 for uridine and 0.7 × 104 s-1, M-1 for cytidine. Our lab has previously shown that UCKL-1 is up-regulated in tumor cells, providing protection against natural killer (NK) cell killing activity. We utilized small interfering RNA (siRNA) to down-regulate UCKL-1 in vitro and in vivo to determine the effect of UCKL-1 on tumor growth and metastasis. The down-regulation of UCKL-1 in YAC-1 lymphoma cells in vitro resulted in decreased cell counts and increased apoptotic activity. Down-regulation of UCKL-1 in K562 leukemia cells in vivo led to decreased primary tumor growth and less tumor cell dissemination and metastasis. These results identify UCKL-1 as a bona fide pyrimidine kinase with the therapeutic potential to be a target for tumor growth inhibition and for diminishing or preventing metastasis.
Collapse
Affiliation(s)
- Emily C. Matchett
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, MO, U.S.A
| | - Elise C. Ambrose
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, MO, U.S.A
| | - Jacki Kornbluth
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, MO, U.S.A
- VA St. Louis Health Care System, St. Louis, MO, U.S.A
| |
Collapse
|
122
|
Donato E, Trumpp A. Targeting the Leukemic stem cell protein machinery by inhibition of mitochondrial pyrimidine synthesis. EMBO Mol Med 2022; 14:e16171. [PMID: 35694783 PMCID: PMC9260207 DOI: 10.15252/emmm.202216171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Acute Myeloid Leukemia is one of the most aggressive blood cancers with a high frequency of relapse. While standard chemotherapy is able to target rapidly proliferating immature blasts, it fails to eradicate slowly proliferating Leukemic Stem Cells. Therefore, new therapeutic strategies that efficiently target LSCs are urgently needed. Recent studies suggest that LSCs have particular metabolic vulnerabilities, which would open the possibility of a therapeutic window with limited off‐target effects on the normal hematopoietic system. In this issue of EMBO Molecular Medicine, So and colleagues investigate the mechanism of action of AG636, a new potent inhibitor of de novo pyrimidine synthesis, and discovered an unexpected link to AML protein translation essential for LSC function.
Collapse
Affiliation(s)
- Elisa Donato
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.,Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.,Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| |
Collapse
|
123
|
Venkatachalam V, Jambhekar A, Lahav G. Reading oscillatory instructions: How cells achieve time-dependent responses to oscillating transcription factors. Curr Opin Cell Biol 2022; 77:102099. [PMID: 35690043 DOI: 10.1016/j.ceb.2022.102099] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/16/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022]
Affiliation(s)
- Veena Venkatachalam
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, 210 Longwood Avenue, Boston, MA 02115, USA; Department of Radiation Oncology, Dana-Farber Brigham Cancer Center, 75 Francis St, Boston, MA 02115, USA
| | - Ashwini Jambhekar
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, 210 Longwood Avenue, Boston, MA 02115, USA; Ludwig Center at Harvard, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02215, USA
| | - Galit Lahav
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, 210 Longwood Avenue, Boston, MA 02115, USA; Ludwig Center at Harvard, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02215, USA.
| |
Collapse
|
124
|
Galati S, Sainas S, Giorgis M, Boschi D, Lolli ML, Ortore G, Poli G, Tuccinardi T. Identification of Human Dihydroorotate Dehydrogenase Inhibitor by a Pharmacophore-Based Virtual Screening Study. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123660. [PMID: 35744791 PMCID: PMC9228440 DOI: 10.3390/molecules27123660] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 11/16/2022]
Abstract
Human dihydroorotate dehydrogenase (hDHODH) is an enzyme belonging to a flavin mononucleotide (FMN)-dependent family involved in de novo pyrimidine biosynthesis, a key biological pathway for highly proliferating cancer cells and pathogens. In fact, hDHODH proved to be a promising therapeutic target for the treatment of acute myelogenous leukemia, multiple myeloma, and viral and bacterial infections; therefore, the identification of novel hDHODH ligands represents a hot topic in medicinal chemistry. In this work, we reported a virtual screening study for the identification of new promising hDHODH inhibitors. A pharmacophore-based approach combined with a consensus docking analysis and molecular dynamics simulations was applied to screen a large database of commercial compounds. The whole virtual screening protocol allowed for the identification of a novel compound that is endowed with promising inhibitory activity against hDHODH and is structurally different from known ligands. These results validated the reliability of the in silico workflow and provided a valuable starting point for hit-to-lead and future lead optimization studies aimed at the development of new potent hDHODH inhibitors.
Collapse
Affiliation(s)
- Salvatore Galati
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (S.G.); (G.O.); (T.T.)
| | - Stefano Sainas
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.S.); (M.G.); (D.B.); (M.L.L.)
| | - Marta Giorgis
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.S.); (M.G.); (D.B.); (M.L.L.)
| | - Donatella Boschi
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.S.); (M.G.); (D.B.); (M.L.L.)
| | - Marco L. Lolli
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.S.); (M.G.); (D.B.); (M.L.L.)
| | - Gabriella Ortore
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (S.G.); (G.O.); (T.T.)
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (S.G.); (G.O.); (T.T.)
- Correspondence: ; Tel.: +39-050-221-9603
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (S.G.); (G.O.); (T.T.)
| |
Collapse
|
125
|
Yang X, Li C, Gou K, Liu X, Zhou Y, Zou J, Chen Q, Luo Y, Zhao Y. A novel and potent dihydroorotate dehydrogenase inhibitor suppresses the proliferation of colorectal cancer by inducing mitochondrial dysfunction and DNA damage. MEDCOMM – ONCOLOGY 2022. [DOI: 10.1002/mog2.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Xiaowei Yang
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant‐Sourced Drug, West China School of Pharmacy, Sichuan Research Center for Drug Precision Industrial Technology Sichuan University Chengdu Sichuan China
| | - Chungen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital West China Medical Chengdu Sichuan China
| | - Kun Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital West China Medical Chengdu Sichuan China
| | - Xiaocong Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital West China Medical Chengdu Sichuan China
| | - Yue Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital West China Medical Chengdu Sichuan China
| | - Jiao Zou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital West China Medical Chengdu Sichuan China
| | - Qiang Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital West China Medical Chengdu Sichuan China
| | - Youfu Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital West China Medical Chengdu Sichuan China
| | - Yinglan Zhao
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant‐Sourced Drug, West China School of Pharmacy, Sichuan Research Center for Drug Precision Industrial Technology Sichuan University Chengdu Sichuan China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital West China Medical Chengdu Sichuan China
| |
Collapse
|
126
|
Pabon CM, Abbas HA, Konopleva M. Acute myeloid leukemia: therapeutic targeting of stem cells. Expert Opin Ther Targets 2022; 26:547-556. [DOI: 10.1080/14728222.2022.2083957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Cindy M. Pabon
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hussein A. Abbas
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Marina Konopleva
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
127
|
Feng X, Tang M, Dede M, Su D, Pei G, Jiang D, Wang C, Chen Z, Li M, Nie L, Xiong Y, Li S, Park JM, Zhang H, Huang M, Szymonowicz K, Zhao Z, Hart T, Chen J. Genome-wide CRISPR screens using isogenic cells reveal vulnerabilities conferred by loss of tumor suppressors. SCIENCE ADVANCES 2022; 8:eabm6638. [PMID: 35559673 PMCID: PMC9106303 DOI: 10.1126/sciadv.abm6638] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 03/30/2022] [Indexed: 05/05/2023]
Abstract
Exploiting cancer vulnerabilities is critical for the discovery of anticancer drugs. However, tumor suppressors cannot be directly targeted because of their loss of function. To uncover specific vulnerabilities for cells with deficiency in any given tumor suppressor(s), we performed genome-scale CRISPR loss-of-function screens using a panel of isogenic knockout cells we generated for 12 common tumor suppressors. Here, we provide a comprehensive and comparative dataset for genetic interactions between the whole-genome protein-coding genes and a panel of tumor suppressor genes, which allows us to uncover known and new high-confidence synthetic lethal interactions. Mining this dataset, we uncover essential paralog gene pairs, which could be a common mechanism for interpreting synthetic lethality. Moreover, we propose that some tumor suppressors could be targeted to suppress proliferation of cells with deficiency in other tumor suppressors. This dataset provides valuable information that can be further exploited for targeted cancer therapy.
Collapse
Affiliation(s)
- Xu Feng
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mengfan Tang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Merve Dede
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dan Su
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Guangsheng Pei
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Dadi Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chao Wang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhen Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mi Li
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Litong Nie
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yun Xiong
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Siting Li
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jeong-Min Park
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Huimin Zhang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Min Huang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Klaudia Szymonowicz
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Traver Hart
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Junjie Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
128
|
So J, Lewis AC, Smith LK, Stanley K, Franich R, Yoannidis D, Pijpers L, Dominguez P, Hogg SJ, Vervoort SJ, Brown FC, Johnstone RW, McDonald G, Ulanet DB, Murtie J, Gruber E, Kats LM. Inhibition of pyrimidine biosynthesis targets protein translation in acute myeloid leukemia. EMBO Mol Med 2022; 14:e15203. [PMID: 35514210 PMCID: PMC9260210 DOI: 10.15252/emmm.202115203] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 12/18/2022] Open
Abstract
The mitochondrial enzyme dihydroorotate dehydrogenase (DHODH) catalyzes one of the rate‐limiting steps in de novo pyrimidine biosynthesis, a pathway that provides essential metabolic precursors for nucleic acids, glycoproteins, and phospholipids. DHODH inhibitors (DHODHi) are clinically used for autoimmune diseases and are emerging as a novel class of anticancer agents, especially in acute myeloid leukemia (AML) where pyrimidine starvation was recently shown to reverse the characteristic differentiation block in AML cells. Herein, we show that DHODH blockade rapidly shuts down protein translation in leukemic stem cells (LSCs) and has potent and selective activity against multiple AML subtypes. Moreover, we find that ablation of CDK5, a gene that is recurrently deleted in AML and related disorders, increases the sensitivity of AML cells to DHODHi. Our studies provide important molecular insights and identify a potential biomarker for an emerging strategy to target AML.
Collapse
Affiliation(s)
- Joan So
- The Peter MacCallum Cancer Centre, Melbourne, Vic., Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic., Australia
| | | | - Lorey K Smith
- The Peter MacCallum Cancer Centre, Melbourne, Vic., Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic., Australia
| | - Kym Stanley
- The Peter MacCallum Cancer Centre, Melbourne, Vic., Australia
| | - Rheana Franich
- The Peter MacCallum Cancer Centre, Melbourne, Vic., Australia
| | - David Yoannidis
- The Peter MacCallum Cancer Centre, Melbourne, Vic., Australia
| | - Lizzy Pijpers
- The Peter MacCallum Cancer Centre, Melbourne, Vic., Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic., Australia
| | - Pilar Dominguez
- The Peter MacCallum Cancer Centre, Melbourne, Vic., Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic., Australia
| | - Simon J Hogg
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stephin J Vervoort
- The Peter MacCallum Cancer Centre, Melbourne, Vic., Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic., Australia
| | - Fiona C Brown
- Australian Centre for Blood Diseases, Monash University, Melbourne, Vic., Australia
| | - Ricky W Johnstone
- The Peter MacCallum Cancer Centre, Melbourne, Vic., Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic., Australia
| | | | | | | | - Emily Gruber
- The Peter MacCallum Cancer Centre, Melbourne, Vic., Australia
| | - Lev M Kats
- The Peter MacCallum Cancer Centre, Melbourne, Vic., Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|
129
|
Wang T, Zhang X, Jia M, Yang A, Liu J, Wen T, Meng J, Xu H. Hydrophilic Realgar Nanocrystals Prolong the Survival of Refractory Acute Myeloid Leukemia Mice Through Inducing Multi-Lineage Differentiation and Apoptosis. Int J Nanomedicine 2022; 17:2191-2202. [PMID: 35599749 PMCID: PMC9122054 DOI: 10.2147/ijn.s358469] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/11/2022] [Indexed: 12/25/2022] Open
Affiliation(s)
- Tao Wang
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Xue Zhang
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Mengfan Jia
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Aiyun Yang
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Jian Liu
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Tao Wen
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Jie Meng
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Haiyan Xu
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
- Correspondence: Haiyan Xu; Jie Meng, Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China, Tel +8610 69156437; +8610 65135502, Email ;
| |
Collapse
|
130
|
Pretreatment of umbilical cord derived MSCs with IFN-γ and TNF-α enhances the tumor-suppressive effect on acute myeloid leukemia. Biochem Pharmacol 2022; 199:115007. [DOI: 10.1016/j.bcp.2022.115007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/09/2022] [Accepted: 03/13/2022] [Indexed: 01/27/2023]
|
131
|
Franklin R, Guo Y, He S, Chen M, Ji F, Zhou X, Frankhouser D, Do BT, Chiem C, Jang M, Blanco MA, Vander Heiden MG, Rockne RC, Ninova M, Sykes DB, Hochedlinger K, Lu R, Sadreyev RI, Murn J, Volk A, Cheloufi S. Regulation of chromatin accessibility by the histone chaperone CAF-1 sustains lineage fidelity. Nat Commun 2022; 13:2350. [PMID: 35487911 PMCID: PMC9054786 DOI: 10.1038/s41467-022-29730-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/25/2022] [Indexed: 12/13/2022] Open
Abstract
Cell fate commitment is driven by dynamic changes in chromatin architecture and activity of lineage-specific transcription factors (TFs). The chromatin assembly factor-1 (CAF-1) is a histone chaperone that regulates chromatin architecture by facilitating nucleosome assembly during DNA replication. Accumulating evidence supports a substantial role of CAF-1 in cell fate maintenance, but the mechanisms by which CAF-1 restricts lineage choice remain poorly understood. Here, we investigate how CAF-1 influences chromatin dynamics and TF activity during lineage differentiation. We show that CAF-1 suppression triggers rapid differentiation of myeloid stem and progenitor cells into a mixed lineage state. We find that CAF-1 sustains lineage fidelity by controlling chromatin accessibility at specific loci, and limiting the binding of ELF1 TF at newly-accessible diverging regulatory elements. Together, our findings decipher key traits of chromatin accessibility that sustain lineage integrity and point to a powerful strategy for dissecting transcriptional circuits central to cell fate commitment.
Collapse
Affiliation(s)
- Reuben Franklin
- Department of Biochemistry, University of California, Riverside, 3401 Watkins Drive, Boyce Hall, Riverside, CA, 92521, United States
- Stem Cell Center, University of California, Riverside, 900 University Ave, Riverside, CA, 92521, United States
| | - Yiming Guo
- Department of Biochemistry, University of California, Riverside, 3401 Watkins Drive, Boyce Hall, Riverside, CA, 92521, United States
- Stem Cell Center, University of California, Riverside, 900 University Ave, Riverside, CA, 92521, United States
| | - Shiyang He
- Department of Biochemistry, University of California, Riverside, 3401 Watkins Drive, Boyce Hall, Riverside, CA, 92521, United States
| | - Meijuan Chen
- Department of Biochemistry, University of California, Riverside, 3401 Watkins Drive, Boyce Hall, Riverside, CA, 92521, United States
- Stem Cell Center, University of California, Riverside, 900 University Ave, Riverside, CA, 92521, United States
| | - Fei Ji
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA, 02114, United States
| | - Xinyue Zhou
- Division of Hematology/Oncology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - David Frankhouser
- Department of Population Sciences City of Hope National Medical Center, Duarte, CA, United States
| | - Brian T Do
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States
- Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA, 02115, United States
| | - Carmen Chiem
- Department of Biochemistry, University of California, Riverside, 3401 Watkins Drive, Boyce Hall, Riverside, CA, 92521, United States
- Stem Cell Center, University of California, Riverside, 900 University Ave, Riverside, CA, 92521, United States
| | - Mihyun Jang
- Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, City of Hope National Medical Center, Duarte, CA, United States
| | - M Andres Blanco
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States
- Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA, 02115, United States
| | - Russell C Rockne
- Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, City of Hope National Medical Center, Duarte, CA, United States
| | - Maria Ninova
- Department of Biochemistry, University of California, Riverside, 3401 Watkins Drive, Boyce Hall, Riverside, CA, 92521, United States
| | - David B Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA, 02114, United States
| | - Konrad Hochedlinger
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA, 02114, United States
- Department of Genetics, Harvard Medical School, 25 Shattuck Street, Boston, MA, 02115, United States
- Harvard Stem Cell Institute, 1350 Massachusetts Avenue, Cambridge, MA, 02138, United States
- Cancer Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA, 02114, United States
| | - Rui Lu
- Division of Hematology/Oncology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ruslan I Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA, 02114, United States
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Jernej Murn
- Department of Biochemistry, University of California, Riverside, 3401 Watkins Drive, Boyce Hall, Riverside, CA, 92521, United States.
| | - Andrew Volk
- Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.
| | - Sihem Cheloufi
- Department of Biochemistry, University of California, Riverside, 3401 Watkins Drive, Boyce Hall, Riverside, CA, 92521, United States.
- Stem Cell Center, University of California, Riverside, 900 University Ave, Riverside, CA, 92521, United States.
| |
Collapse
|
132
|
Targeting nucleotide metabolism: a promising approach to enhance cancer immunotherapy. J Hematol Oncol 2022; 15:45. [PMID: 35477416 PMCID: PMC9044757 DOI: 10.1186/s13045-022-01263-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/18/2022] [Indexed: 12/12/2022] Open
Abstract
Targeting nucleotide metabolism can not only inhibit tumor initiation and progression but also exert serious side effects. With in-depth studies of nucleotide metabolism, our understanding of nucleotide metabolism in tumors has revealed their non-proliferative effects on immune escape, indicating the potential effectiveness of nucleotide antimetabolites for enhancing immunotherapy. A growing body of evidence now supports the concept that targeting nucleotide metabolism can increase the antitumor immune response by (1) activating host immune systems via maintaining the concentrations of several important metabolites, such as adenosine and ATP, (2) promoting immunogenicity caused by increased mutability and genomic instability by disrupting the purine and pyrimidine pool, and (3) releasing nucleoside analogs via microbes to regulate immunity. Therapeutic approaches targeting nucleotide metabolism combined with immunotherapy have achieved exciting success in preclinical animal models. Here, we review how dysregulated nucleotide metabolism can promote tumor growth and interact with the host immune system, and we provide future insights into targeting nucleotide metabolism for immunotherapeutic treatment of various malignancies.
Collapse
|
133
|
Apaolaza I, San José-Enériz E, Valcarcel LV, Agirre X, Prosper F, Planes FJ. A network-based approach to integrate nutrient microenvironment in the prediction of synthetic lethality in cancer metabolism. PLoS Comput Biol 2022; 18:e1009395. [PMID: 35286311 PMCID: PMC8947600 DOI: 10.1371/journal.pcbi.1009395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 03/24/2022] [Accepted: 02/18/2022] [Indexed: 11/18/2022] Open
Abstract
Synthetic Lethality (SL) is currently defined as a type of genetic interaction in which the loss of function of either of two genes individually has limited effect in cell viability but inactivation of both genes simultaneously leads to cell death. Given the profound genomic aberrations acquired by tumor cells, which can be systematically identified with -omics data, SL is a promising concept in cancer research. In particular, SL has received much attention in the area of cancer metabolism, due to the fact that relevant functional alterations concentrate on key metabolic pathways that promote cellular proliferation. With the extensive prior knowledge about human metabolic networks, a number of computational methods have been developed to predict SL in cancer metabolism, including the genetic Minimal Cut Sets (gMCSs) approach. A major challenge in the application of SL approaches to cancer metabolism is to systematically integrate tumor microenvironment, given that genetic interactions and nutritional availability are interconnected to support proliferation. Here, we propose a more general definition of SL for cancer metabolism that combines genetic and environmental interactions, namely loss of gene functions and absence of nutrients in the environment. We extend our gMCSs approach to determine this new family of metabolic synthetic lethal interactions. A computational and experimental proof-of-concept is presented for predicting the lethality of dihydrofolate reductase (DHFR) inhibition in different environments. Finally, our approach is applied to identify extracellular nutrient dependences of tumor cells, elucidating cholesterol and myo-inositol depletion as potential vulnerabilities in different malignancies. Metabolic reprogramming is one of the hallmarks of tumor cells. Synthetic lethality (SL) is a promising approach to exploit these metabolic alterations and elucidate cancer-specific genetic dependences. However, current SL approaches do not systematically consider tumor microenvironment, which is particularly important in cancer metabolism in order to generalize identified genetic dependences. In this article, we directly address this issue and propose a more general approach to SL that integrates both genetic and environmental context of tumor cells. Our definition can help to contextualize genetic dependencies in different environmental scenarios, but it could also reveal nutrient dependencies according to the genetic context. We also provide a computational pipeline to identify this new family of synthetic lethals in genome-scale metabolic networks. A computational and experimental proof-of-concept is presented for predicting the lethality of dihydrofolate reductase (DHFR) inhibition in different environments. Finally, our approach is applied to identify extracellular nutrient dependences of cancer cell lines, elucidating cholesterol and myo-Inositol depletion as potential vulnerabilities in different malignancies.
Collapse
Affiliation(s)
- Iñigo Apaolaza
- Universidad de Navarra, Tecnun Escuela de Ingeniería, San Sebastián, Spain
- Universidad de Navarra, Centro de Ingeniería Biomédica, Pamplona, Spain
- Universidad de Navarra, DATAI Instituto de Ciencia de los Datos e Inteligencia Artificial, Pamplona, Spain
| | - Edurne San José-Enériz
- Universidad de Navarra, CIMA Centro de Investigación de Medicina Aplicada, Pamplona, Spain
- IdiSNA Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain
| | - Luis V. Valcarcel
- Universidad de Navarra, Tecnun Escuela de Ingeniería, San Sebastián, Spain
- Universidad de Navarra, CIMA Centro de Investigación de Medicina Aplicada, Pamplona, Spain
- IdiSNA Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Xabier Agirre
- Universidad de Navarra, CIMA Centro de Investigación de Medicina Aplicada, Pamplona, Spain
- IdiSNA Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain
| | - Felipe Prosper
- Universidad de Navarra, CIMA Centro de Investigación de Medicina Aplicada, Pamplona, Spain
- IdiSNA Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain
- Clínica Universidad de Navarra, Pamplona, Spain
| | - Francisco J. Planes
- Universidad de Navarra, Tecnun Escuela de Ingeniería, San Sebastián, Spain
- Universidad de Navarra, Centro de Ingeniería Biomédica, Pamplona, Spain
- Universidad de Navarra, DATAI Instituto de Ciencia de los Datos e Inteligencia Artificial, Pamplona, Spain
- * E-mail:
| |
Collapse
|
134
|
Xiang W, Lam YH, Periyasamy G, Chuah C. Application of High Throughput Technologies in the Development of Acute Myeloid Leukemia Therapy: Challenges and Progress. Int J Mol Sci 2022; 23:ijms23052863. [PMID: 35270002 PMCID: PMC8910862 DOI: 10.3390/ijms23052863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 11/27/2022] Open
Abstract
Acute myeloid leukemia (AML) is a complex hematological malignancy characterized by extensive heterogeneity in genetics, response to therapy and long-term outcomes, making it a prototype example of development for personalized medicine. Given the accessibility to hematologic malignancy patient samples and recent advances in high-throughput technologies, large amounts of biological data that are clinically relevant for diagnosis, risk stratification and targeted drug development have been generated. Recent studies highlight the potential of implementing genomic-based and phenotypic-based screens in clinics to improve survival in patients with refractory AML. In this review, we will discuss successful applications as well as challenges of most up-to-date high-throughput technologies, including artificial intelligence (AI) approaches, in the development of personalized medicine for AML, and recent clinical studies for evaluating the utility of integrating genomics-guided and drug sensitivity testing-guided treatment approaches for AML patients.
Collapse
Affiliation(s)
- Wei Xiang
- Department of Haematology, Singapore General Hospital, Singapore 169608, Singapore; (W.X.); (Y.H.L.)
| | - Yi Hui Lam
- Department of Haematology, Singapore General Hospital, Singapore 169608, Singapore; (W.X.); (Y.H.L.)
| | - Giridharan Periyasamy
- High Throughput Phenomics Platform, Experimental Drug Development Centre, Agency for Science, Technology and Research (A*STAR), Singapore 139632, Singapore;
| | - Charles Chuah
- Department of Haematology, Singapore General Hospital, Singapore 169608, Singapore; (W.X.); (Y.H.L.)
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Correspondence:
| |
Collapse
|
135
|
Branstrom A, Cao L, Furia B, Trotta C, Santaguida M, Graci JD, Colacino JM, Ray B, Li W, Sheedy J, Mollin A, Yeh S, Kong R, Sheridan R, Baird JD, O'Keefe K, Spiegel R, Goodwin E, Keating S, Weetall M. Emvododstat, a Potent Dihydroorotate Dehydrogenase Inhibitor, Is Effective in Preclinical Models of Acute Myeloid Leukemia. Front Oncol 2022; 12:832816. [PMID: 35223511 PMCID: PMC8864546 DOI: 10.3389/fonc.2022.832816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
Blocking the pyrimidine nucleotide de novo synthesis pathway by inhibiting dihydroorotate dehydrogenase (DHODH) results in the cell cycle arrest and/or differentiation of rapidly proliferating cells including activated lymphocytes, cancer cells, or virally infected cells. Emvododstat (PTC299) is an orally bioavailable small molecule that inhibits DHODH. We evaluated the potential for emvododstat to inhibit the progression of acute myeloid leukemia (AML) using several in vitro and in vivo models of the disease. Broad potent activity was demonstrated against multiple AML cell lines, AML blasts cultured ex vivo from patient blood samples, and AML tumor models including patient-derived xenograft models. Emvododstat induced differentiation, cytotoxicity, or both in primary AML patient blasts cultured ex vivo with 8 of 10 samples showing sensitivity. AML cells with diverse driver mutations were sensitive, suggesting the potential of emvododstat for broad therapeutic application. AML cell lines that are not sensitive to emvododstat are likely to be more reliant on the salvage pathway than on de novo synthesis of pyrimidine nucleotides. Pharmacokinetic experiments in rhesus monkeys demonstrated that emvododstat levels rose rapidly after oral administration, peaking about 2 hours post-dosing. This was associated with an increase in the levels of dihydroorotate (DHO), the substrate for DHODH, within 2 hours of dosing indicating that DHODH inhibition is rapid. DHO levels declined as drug levels declined, consistent with the reversibility of DHODH inhibition by emvododstat. These preclinical findings provide a rationale for clinical evaluation of emvododstat in an ongoing Phase 1 study of patients with relapsed/refractory acute leukemias.
Collapse
Affiliation(s)
- Arthur Branstrom
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Liangxian Cao
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Bansri Furia
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | | | | | - Jason D Graci
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Joseph M Colacino
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Balmiki Ray
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Wencheng Li
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Josephine Sheedy
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Anna Mollin
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Shirley Yeh
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Ronald Kong
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | | | - John D Baird
- Clinical, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Kylie O'Keefe
- Commercial, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Robert Spiegel
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Elizabeth Goodwin
- Scientific Writing, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Suzanne Keating
- Scientific Writing, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Marla Weetall
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| |
Collapse
|
136
|
Mesbahi Y, Trahair TN, Lock RB, Connerty P. Exploring the Metabolic Landscape of AML: From Haematopoietic Stem Cells to Myeloblasts and Leukaemic Stem Cells. Front Oncol 2022; 12:807266. [PMID: 35223487 PMCID: PMC8867093 DOI: 10.3389/fonc.2022.807266] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/18/2022] [Indexed: 12/13/2022] Open
Abstract
Despite intensive chemotherapy regimens, up to 60% of adults with acute myeloid leukaemia (AML) will relapse and eventually succumb to their disease. Recent studies suggest that leukaemic stem cells (LSCs) drive AML relapse by residing in the bone marrow niche and adapting their metabolic profile. Metabolic adaptation and LSC plasticity are novel hallmarks of leukemogenesis that provide important biological processes required for tumour initiation, progression and therapeutic responses. These findings highlight the importance of targeting metabolic pathways in leukaemia biology which might serve as the Achilles' heel for the treatment of AML relapse. In this review, we highlight the metabolic differences between normal haematopoietic cells, bulk AML cells and LSCs. Specifically, we focus on four major metabolic pathways dysregulated in AML; (i) glycolysis; (ii) mitochondrial metabolism; (iii) amino acid metabolism; and (iv) lipid metabolism. We then outline established and emerging drug interventions that exploit metabolic dependencies of leukaemic cells in the treatment of AML. The metabolic signature of AML cells alters during different biological conditions such as chemotherapy and quiescence. Therefore, targeting the metabolic vulnerabilities of these cells might selectively eradicate them and improve the overall survival of patients with AML.
Collapse
Affiliation(s)
- Yashar Mesbahi
- Children's Cancer Institute, Lowy Cancer Centre, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,University of New South Wales Centre for Childhood Cancer Research, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia
| | - Toby N Trahair
- Children's Cancer Institute, Lowy Cancer Centre, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Centre, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,University of New South Wales Centre for Childhood Cancer Research, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia
| | - Patrick Connerty
- Children's Cancer Institute, Lowy Cancer Centre, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,University of New South Wales Centre for Childhood Cancer Research, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia
| |
Collapse
|
137
|
Huang K, Matsumura H, Zhao Y, Herbig M, Yuan D, Mineharu Y, Harmon J, Findinier J, Yamagishi M, Ohnuki S, Nitta N, Grossman AR, Ohya Y, Mikami H, Isozaki A, Goda K. Deep imaging flow cytometry. LAB ON A CHIP 2022; 22:876-889. [PMID: 35142325 DOI: 10.1039/d1lc01043c] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Imaging flow cytometry (IFC) has become a powerful tool for diverse biomedical applications by virtue of its ability to image single cells in a high-throughput manner. However, there remains a challenge posed by the fundamental trade-off between throughput, sensitivity, and spatial resolution. Here we present deep-learning-enhanced imaging flow cytometry (dIFC) that circumvents this trade-off by implementing an image restoration algorithm on a virtual-freezing fluorescence imaging (VIFFI) flow cytometry platform, enabling higher throughput without sacrificing sensitivity and spatial resolution. A key component of dIFC is a high-resolution (HR) image generator that synthesizes "virtual" HR images from the corresponding low-resolution (LR) images acquired with a low-magnification lens (10×/0.4-NA). For IFC, a low-magnification lens is favorable because of reduced image blur of cells flowing at a higher speed, which allows higher throughput. We trained and developed the HR image generator with an architecture containing two generative adversarial networks (GANs). Furthermore, we developed dIFC as a method by combining the trained generator and IFC. We characterized dIFC using Chlamydomonas reinhardtii cell images, fluorescence in situ hybridization (FISH) images of Jurkat cells, and Saccharomyces cerevisiae (budding yeast) cell images, showing high similarities of dIFC images to images obtained with a high-magnification lens (40×/0.95-NA), at a high flow speed of 2 m s-1. We lastly employed dIFC to show enhancements in the accuracy of FISH-spot counting and neck-width measurement of budding yeast cells. These results pave the way for statistical analysis of cells with high-dimensional spatial information.
Collapse
Affiliation(s)
- Kangrui Huang
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Hiroki Matsumura
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Yaqi Zhao
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Maik Herbig
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Dan Yuan
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Yohei Mineharu
- Department of Neurosurgery, Kyoto University, Kyoto 606-8507, Japan
- Department of Artificial Intelligence in Healthcare and Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Jeffrey Harmon
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Justin Findinier
- Department of Plant Biology, The Carnegie Institution for Science, Stanford, California 94305, USA
| | - Mai Yamagishi
- Department of Biological Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Shinsuke Ohnuki
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8562, Japan
| | | | - Arthur R Grossman
- Department of Plant Biology, The Carnegie Institution for Science, Stanford, California 94305, USA
- Department of Biology, Stanford University, Stanford, California 94305, USA
| | - Yoshikazu Ohya
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8562, Japan
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Tokyo 113-8654, Japan
| | - Hideharu Mikami
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
- PRESTO, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Akihiro Isozaki
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Keisuke Goda
- Department of Chemistry, The University of Tokyo, Tokyo 113-0033, Japan.
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
- Institute of Technological Sciences, Wuhan University, Hubei 430072, China
| |
Collapse
|
138
|
New Insights into the Interaction of Class II Dihydroorotate Dehydrogenases with Ubiquinone in Lipid Bilayers as a Function of Lipid Composition. Int J Mol Sci 2022; 23:ijms23052437. [PMID: 35269583 PMCID: PMC8910288 DOI: 10.3390/ijms23052437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 12/10/2022] Open
Abstract
The fourth enzymatic reaction in the de novo pyrimidine biosynthesis, the oxidation of dihydroorotate to orotate, is catalyzed by dihydroorotate dehydrogenase (DHODH). Enzymes belonging to the DHODH Class II are membrane-bound proteins that use ubiquinones as their electron acceptors. We have designed this study to understand the interaction of an N-terminally truncated human DHODH (HsΔ29DHODH) and the DHODH from Escherichia coli (EcDHODH) with ubiquinone (Q10) in supported lipid membranes using neutron reflectometry (NR). NR has allowed us to determine in situ, under solution conditions, how the enzymes bind to lipid membranes and to unambiguously resolve the location of Q10. Q10 is exclusively located at the center of all of the lipid bilayers investigated, and upon binding, both of the DHODHs penetrate into the hydrophobic region of the outer lipid leaflet towards the Q10. We therefore show that the interaction between the soluble enzymes and the membrane-embedded Q10 is mediated by enzyme penetration. We can also show that EcDHODH binds more efficiently to the surface of simple bilayers consisting of 1-palmitoyl, 2-oleoyl phosphatidylcholine, and tetraoleoyl cardiolipin than HsΔ29DHODH, but does not penetrate into the lipids to the same degree. Our results also highlight the importance of Q10, as well as lipid composition, on enzyme binding.
Collapse
|
139
|
Walter M, Herr P. Re-Discovery of Pyrimidine Salvage as Target in Cancer Therapy. Cells 2022; 11:cells11040739. [PMID: 35203388 PMCID: PMC8870348 DOI: 10.3390/cells11040739] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/10/2022] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
Nucleotides are synthesized through two distinct pathways: de novo synthesis and nucleoside salvage. Whereas the de novo pathway synthesizes nucleotides from amino acids and glucose, the salvage pathway recovers nucleosides or bases formed during DNA or RNA degradation. In contrast to high proliferating non-malignant cells, which are highly dependent on the de novo synthesis, cancer cells can switch to the nucleoside salvage pathways to maintain efficient DNA replication. Pyrimidine de novo synthesis remains the target of interest in cancer therapy and several inhibitors showed promising results in cancer cells and in vivo models. In the 1980s and 1990s, poor responses were however observed in clinical trials with several of the currently existing pyrimidine synthesis inhibitors. To overcome the observed limitations in clinical trials, targeting pyrimidine salvage alone or in combination with pyrimidine de novo inhibitors was suggested. Even though this approach showed initially promising results, it received fresh attention only recently. Here we discuss the re-discovery of targeting pyrimidine salvage pathways for DNA replication alone or in combination with inhibitors of pyrimidine de novo synthesis to overcome limitations of commonly used antimetabolites in various preclinical cancer models and clinical trials. We also highlight newly emerged targets in pyrimidine synthesis as well as pyrimidine salvage as a promising target in immunotherapy.
Collapse
|
140
|
Li Z, Zhang J, Zhou M, Li JL, Qiu QC, Fu JH, Xue SL, Qiu HY. Epigenetic therapy with chidamide alone or combined with 5‑azacitidine exerts antitumour effects on acute myeloid leukaemia cells in vitro. Oncol Rep 2022; 47:66. [PMID: 35103292 PMCID: PMC8848469 DOI: 10.3892/or.2022.8277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/04/2022] [Indexed: 11/06/2022] Open
Abstract
Chidamide, a selective histone deacetylase inhibitor, has antitumour effects. 5‑azacitidine (5‑AZA), a hypomethylating agent, is effective in treating acute myeloid leukaemia (AML) and myelodysplastic syndrome. However, to the best of our knowledge, the effect of chidamide and 5‑AZA on AML cell lines has not been fully investigated. In the present study, the antileukaemia activity of chidamide, alone and in combination with 5‑AZA, was assessed on different subtypes of AML cell lines (M1‑M5) and primary samples from several patients with AML in vitro. The results indicated that the proliferation of leukaemia cells was significantly and dose‑dependently inhibited by chidamide and 5‑AZA alone or in combination. The combination also had marked synergistic effects to induce apoptosis of AML cells. The apoptosis of leukaemia cells was induced via downregulation of BCL‑2 and myeloid‑cell leukemia 1 (MCL‑1) levels. Of note, chidamide also degraded the MCL‑1 protein in venetoclax‑resistant U937 cells, in which the MCL‑1 protein is upregulated. In addition, chidamide was able to induce myeloid differentiation (with CD11b upregulation) of AML cell lines or monocytic/dendritic differentiation (with CD86 upregulation) of primary cultured cells from several patients with AML. Chidamide was also able to promote the differentiation of the venetoclax‑resistant U937 cell line by upregulating CD11b expression. In conclusion, chidamide alone or combined with 5‑AZA may be an effective therapy for AML.
Collapse
Affiliation(s)
- Zheng Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jian Zhang
- Department of Blood Transfusion, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Min Zhou
- Department of Hematology, Changshu Affiliated Hospital of Soochow University, The First People's Hospital of Changshu, Changshu, Jiangsu 215500, P.R. China
| | - Jin-Li Li
- State Key Laboratory of Radiation Medicine and Protection, Oncology Radiotherapy Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Qiao-Cheng Qiu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jian-Hong Fu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Sheng-Li Xue
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Hui-Ying Qiu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
141
|
Bedi M, Ray M, Ghosh A. Active mitochondrial respiration in cancer: a target for the drug. Mol Cell Biochem 2022; 477:345-361. [PMID: 34716860 DOI: 10.1007/s11010-021-04281-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 10/21/2021] [Indexed: 12/21/2022]
Abstract
The relative contribution of mitochondrial respiration and subsequent energy production in malignant cells has remained controversial to date. Enhanced aerobic glycolysis and impaired mitochondrial respiration have gained more attention in the metabolic study of cancer. In contrast to the popular concept, mitochondria of cancer cells oxidize a diverse array of metabolic fuels to generate a majority of the cellular energy by respiration. Several mitochondrial respiratory chain (MRC) subunits' expressions are critical for the growth, metastasis, and cancer cell invasion. Also, the assembly factors, which regulate the integration of individual MRC complexes into native super-complexes, are upregulated in cancer. Moreover, a series of anti-cancer drugs function by inhibiting respiration and ATP production. In this review, we have specified the roles of mitochondrial fuels, MRC subunits, and super-complex assembly factors that promote active respiration across different cancer types and discussed the potential roles of MRC inhibitor drugs in controlling cancer.
Collapse
Affiliation(s)
- Minakshi Bedi
- Department of Biochemistry, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, West Bengal, 700019, India
| | - Manju Ray
- Department of Biophysics, Bose Institute, P 1/12, CIT Scheme VII M, Kolkata, West Bengal, 700054, India
- Department of Chemistry, Institute of Applied Science & Humanities GLA University Mathura, 17km Stone, NH-2, Mathura-Delhi Road, Mathura, UP, 281 406, India
| | - Alok Ghosh
- Department of Biochemistry, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, West Bengal, 700019, India.
| |
Collapse
|
142
|
Zhang L, Zhang J, Wang J, Ren C, Tang P, Ouyang L, Wang Y. Recent advances of human dihydroorotate dehydrogenase inhibitors for cancer therapy: Current development and future perspectives. Eur J Med Chem 2022; 232:114176. [DOI: 10.1016/j.ejmech.2022.114176] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/19/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
|
143
|
Orozco Rodriguez JM, Krupinska E, Wacklin-Knecht H, Knecht W. Protein production, kinetic and biophysical characterization of three human dihydroorotate dehydrogenase mutants associated with Miller syndrome. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2022; 41:1318-1336. [PMID: 35094635 DOI: 10.1080/15257770.2021.2023749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Miller syndrome is a rare Mendelian disorder caused by mutations in the gene encoding human dihydroorotate dehydrogenase (DHODH). Human DHODH, a Class II DHODH, is an integral protein of the inner mitochondrial membrane (IMM) catalyzing the fourth step of the de novo pyrimidine biosynthesis pathway. Here we present a summary of the state of knowledge regarding Miller syndrome in the absence of any current review on the topic. We then describe the production and characterization of three distinct DHODH missense mutations (G19E, E52G, R135C) associated with Miller syndrome by means of enzyme kinetics and biophysical techniques. These human DHODH mutants were produced both in E. coli and in insect cells using the baculovirus expression vector system. We can show that the effects of these mutations differ from each other and the wild-type enzyme with respect to decreased enzymatic activity, decreased protein stability and probably disturbance of the correct import into the IMM. In addition, our results show that the N-terminus of human DHODH is not only a structural element necessary for correct mitochondrial import and location of DHODH on the outer side of the IMM, but also influences thermal stability, enzymatic activity and affects the kinetic parameters.Supplemental data for this article is available online at https://doi.org/10.1080/15257770.2021.2023749 .
Collapse
Affiliation(s)
| | - Ewa Krupinska
- Department of Biology & Lund Protein Production Platform, Lund University, Lund, Sweden
| | - Hanna Wacklin-Knecht
- Department of Chemistry, Division of Physical Chemistry, Lund University, Lund, Sweden.,European Spallation Source ERIC, Lund, Sweden
| | - Wolfgang Knecht
- Department of Biology & Lund Protein Production Platform, Lund University, Lund, Sweden
| |
Collapse
|
144
|
Man CH, Mercier FE, Liu N, Dong W, Stephanopoulos G, Jiang L, Jung Y, Lin CP, Leung AYH, Scadden DT. Proton export alkalinizes intracellular pH and reprograms carbon metabolism to drive normal and malignant cell growth. Blood 2022; 139:502-522. [PMID: 34610101 PMCID: PMC8796654 DOI: 10.1182/blood.2021011563] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 09/20/2021] [Indexed: 01/29/2023] Open
Abstract
Proton export is often considered a detoxifying process in animal cells, with monocarboxylate symporters coexporting excessive lactate and protons during glycolysis or the Warburg effect. We report a novel mechanism by which lactate/H+ export is sufficient to induce cell growth. Increased intracellular pH selectively activates catalysis by key metabolic gatekeeper enzymes HK1/PKM2/G6PDH, thereby enhancing glycolytic and pentose phosphate pathway carbon flux. The result is increased nucleotide levels, NADPH/NADP+ ratio, and cell proliferation. Simply increasing the lactate/proton symporter monocarboxylate transporter 4 (MCT4) or the sodium-proton antiporter NHE1 was sufficient to increase intracellular pH and give normal hematopoietic cells a significant competitive growth advantage in vivo. This process does not require additional cytokine triggers and is exploited in malignancy, where leukemogenic mutations epigenetically increase MCT4. Inhibiting MCT4 decreased intracellular pH and carbon flux and eliminated acute myeloid leukemia-initiating cells in mice without cytotoxic chemotherapy. Intracellular alkalization is a primitive mechanism by which proton partitioning can directly reprogram carbon metabolism for cell growth.
Collapse
Affiliation(s)
- Cheuk Him Man
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Ludwig Center, Harvard Medical School, Boston, MA
| | - Francois E Mercier
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Ludwig Center, Harvard Medical School, Boston, MA
| | - Nian Liu
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Wentao Dong
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Gregory Stephanopoulos
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Li Jiang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA
| | - Yookyung Jung
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA; and
| | - Charles P Lin
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA; and
| | - Anskar Y H Leung
- Division of Haematology, Department of Medicine, University of Hong Kong, Pok Fu Lam, Hong Kong SAR
| | - David T Scadden
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Ludwig Center, Harvard Medical School, Boston, MA
| |
Collapse
|
145
|
Wang H, He X, Li Z, Jin H, Wang X, Li L. Guanosine primes acute myeloid leukemia for differentiation via guanine nucleotide salvage synthesis. Am J Cancer Res 2022; 12:427-444. [PMID: 35141027 PMCID: PMC8822274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/27/2021] [Indexed: 06/14/2023] Open
Abstract
Differentiation arrest represents a distinct hallmark of acute myeloid leukemia (AML). Identification of differentiation-induction agents that are effective across various subtypes remains an unmet challenge. GTP biosynthesis is elevated in several types of cancers, considered to support uncontrolled tumor growth. Here we report that GTP overload by supplementation of guanosine, the nucleoside precursor of GTP, poises AML cells for differentiation and growth inhibition. Transcriptome profiling of guanosine-treated AML cells reveals a myeloid differentiation pattern. Importantly, the treatment compromises leukemia progression in AML xenograft models. Mechanistically, GTP overproduction requires sequential metabolic conversions executed by the purine salvage biosynthesis pathway including the involvement of purine nucleoside phosphorylase (PNP) and hypoxanthine phosphoribosyltransferase 1 (HPRT1). Taken together, our study offers novel metabolic insights tethering GTP homeostasis to myeloid differentiation and provides an experimental basis for further clinical investigations of guanosine or guanine nucleotides in the treatment of AML patients.
Collapse
Affiliation(s)
- Hanying Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou 310016, Zhejiang, China
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Beckman Research Institute, City of Hope National Medical CenterDuarte, CA 91010, USA
| | - Xin He
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Beckman Research Institute, City of Hope National Medical CenterDuarte, CA 91010, USA
| | - Zheng Li
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Beckman Research Institute, City of Hope National Medical CenterDuarte, CA 91010, USA
| | - Hongchuan Jin
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang Province, Cancer Center of Zhejiang University, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou 310016, Zhejiang, China
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou 310016, Zhejiang, China
| | - Ling Li
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Beckman Research Institute, City of Hope National Medical CenterDuarte, CA 91010, USA
| |
Collapse
|
146
|
Miyake S, Masuda S. Inhibition of mitochondrial complex III or dihydroorotate dehydrogenase (DHODH) triggers formation of poly(A) + RNA foci adjacent to nuclear speckles following activation of ATM (ataxia telangiectasia mutated). RNA Biol 2022; 19:1244-1255. [PMID: 36412986 PMCID: PMC9683070 DOI: 10.1080/15476286.2022.2146919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Intracellular and intercellular signalling networks play an essential role in optimizing cellular homoeostasis and are thought to be partly reflected in nuclear mRNA dynamics. However, the regulation of nuclear mRNA dynamics by intracellular and intercellular signals remains largely unexplored, and research tools are lacking. Through an original screening based on the mRNA metabolic mechanism, we discovered that eight well-known inhibitors cause significant nuclear poly(A)+ RNA accumulation. Among these inhibitors, we discovered a new mRNA metabolic response in which the addition of antimycin A, an inhibitor of mitochondrial respiratory-chain complex III (complex III), resulted in a marked accumulation of poly(A)+ RNA near the nuclear speckles. Furthermore, dihydroorotate dehydrogenase (DHODH) inhibitors, a rate-limiting enzyme in the intracellular de novo pyrimidine synthesis reaction that specifically exchanges electrons with complex III, also caused a remarkable accumulation of nuclear poly(A)+ RNA adjacent to the nuclear speckles, which was abolished by extracellular uridine supply, indicating that the depletion of intracellular pyrimidine affects poly(A)+ RNA metabolism. Further analysis revealed that ataxia telangiectasia mutated (ATM), a serine and threonine kinase and a master regulator of DNA double-strand break (DSB) and nucleolar stress, is required for this poly(A)+ RNA nuclear accumulation phenomenon. This study reports new insights into novel aspects of nuclear poly(A)+ RNA metabolism, especially the relationship between mitochondrial respiratory-chain functions, pyrimidine metabolism, and nuclear RNA metabolism.
Collapse
Affiliation(s)
- Shuntaro Miyake
- Division of Integrated Life Sciences, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Seiji Masuda
- Division of Integrated Life Sciences, Graduate School of Biostudies, Kyoto University, Kyoto, Japan,Department of Food Science and Nutrition, Faculty of Agriculture, Kindai University, Nara, Japan,Agricultural Technology and Innovation Research Institute, Kindai University, Nara, Japan,Antiaging Center, Kindai University, Higashiosaka, Japan,CONTACT Seiji Masuda Department of Food Science and Nutrition, Faculty of Agriculture, Kindai University, Nara631-8505, Japan
| |
Collapse
|
147
|
Sireesha R, Tej MB, Poojith N, Sreenivasulu R, Musuluri M, Subbarao M. Synthesis of Substituted Aryl Incorporated Oxazolo[4,5-b]Pyridine-Triazole Derivatives: Anticancer Evaluation and Molecular Docking Studies. Polycycl Aromat Compd 2021. [DOI: 10.1080/10406638.2021.2021256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Reddymasu Sireesha
- Department of Chemistry, Acharya Nagarjuna University, Guntur, Andhra Pradesh, India
| | - Mandava Bhuvan Tej
- Department of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamilnadu, India
| | | | - Reddymasu Sreenivasulu
- Department of Chemistry, University College of Engineering (Autonomous), Jawaharlal Nehru Technological University, Kakinada, Andhra Pradesh, India
| | - Murali Musuluri
- Department of Chemistry, RVR & JC College of Engineering, Guntur, Andhra Pradesh, India
| | - Mannam Subbarao
- Department of Chemistry, Acharya Nagarjuna University, Guntur, Andhra Pradesh, India
| |
Collapse
|
148
|
Chen JA, Ma H, Liu Z, Tian J, Lu S, Fang W, Ze S, Lu W, Xie Q, Huang J, Wang Y. Discovery of Orally Available Retinoic Acid Receptor-Related Orphan Receptor γ-t/Dihydroorotate Dehydrogenase Dual Inhibitors for the Treatment of Refractory Inflammatory Bowel Disease. J Med Chem 2021; 65:592-615. [PMID: 34957834 DOI: 10.1021/acs.jmedchem.1c01746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Inflammatory bowel disease (IBD) is a multifactorial autoimmune disease, representing a major clinical challenge. Herein, a strategy of dual-targeting approach employing retinoic acid receptor-related orphan receptor γ-t (RORγt) and dihydroorotate dehydrogenase (DHODH) was proposed for the treatment of IBD. Dual RORγt/DHODH inhibitors are expected not only to reduce RORγt-driven Th17 cell differentiation but also to mitigate the expansion and activation of T cells, which may enhance anti-inflammatory effects. Starting from 2-aminobenzothiazole hit 1, a series of 2-aminotetrahydrobenzothiazoles were discovered as potent dual RORγt/DHODH inhibitors. Compound 14d stands out with IC50 values of 0.110 μM for RORγt and of 0.297 μM for DHODH. With acceptable mouse pharmacokinetic profiles, 14d exhibited remarkable in vivo anti-inflammatory activity and dose-dependently alleviated the severity of dextran sulfate sodium (DSS)-induced acute colitis in mice. Taken together, the present study provides a novel framework for the development of therapeutic agents for the treatment of IBD.
Collapse
Affiliation(s)
- Ji-An Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Hui Ma
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Zehui Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Jinlong Tian
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Sisi Lu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Wenqing Fang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Shuyin Ze
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Qiong Xie
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China.,Fudan Zhangjiang Institute, 666 Zhangheng Road, Shanghai 201203, China
| | - Jin Huang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yonghui Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| |
Collapse
|
149
|
Li C, Yang X, Luo Y, Liu H, Zhong X, Zhou X, Zeng T, Tao L, Zhou Y, Gou K, Yang X, Liu X, Chen Q, Zhao Y, Luo Y. Design, Synthesis, and Biological Evaluation of a Novel Series of Teriflunomide Derivatives as Potent Human Dihydroorotate Dehydrogenase Inhibitors for Malignancy Treatment. J Med Chem 2021; 64:18175-18192. [PMID: 34905371 DOI: 10.1021/acs.jmedchem.1c01711] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Human dihydroorotate dehydrogenase (hDHODH), as the fourth and rate-limiting enzyme of the de novo pyrimidine synthesis pathway, is regarded as an attractive target for malignancy therapy. In the present study, a novel series of teriflunomide derivatives were designed, synthesized, and evaluated as hDHODH inhibitors. 13t was the optimal compound with promising enzymatic activity (IC50 = 16.0 nM), potent antiproliferative activity against human lymphoma Raji cells (IC50 = 7.7 nM), and excellent aqueous solubility (20.1 mg/mL). Mechanistically, 13t directly inhibited hDHODH and induced cell cycle S-phase arrest in Raji cells. The acute toxicity assay indicated a favorable safety profile of 13t. Notably, 13t displayed significant tumor growth inhibition activity with a tumor growth inhibition (TGI) rate of 81.4% at 30 mg/kg in a Raji xenograft model. Together, 13t is a promising inhibitor of hDHODH and a preclinical candidate for antitumor therapy, especially for lymphoma.
Collapse
Affiliation(s)
- Chungen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Xiaowei Yang
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yuan Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Huan Liu
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xi Zhong
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xia Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Ting Zeng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Lei Tao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Yue Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Kun Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Xinyu Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Xiaocong Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Qiang Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Yinglan Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China.,Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Youfu Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| |
Collapse
|
150
|
Pemovska T, Bigenzahn JW, Srndic I, Lercher A, Bergthaler A, César-Razquin A, Kartnig F, Kornauth C, Valent P, Staber PB, Superti-Furga G. Metabolic drug survey highlights cancer cell dependencies and vulnerabilities. Nat Commun 2021; 12:7190. [PMID: 34907165 PMCID: PMC8671470 DOI: 10.1038/s41467-021-27329-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/16/2021] [Indexed: 12/15/2022] Open
Abstract
Interrogation of cellular metabolism with high-throughput screening approaches can unravel contextual biology and identify cancer-specific metabolic vulnerabilities. To systematically study the consequences of distinct metabolic perturbations, we assemble a comprehensive metabolic drug library (CeMM Library of Metabolic Drugs; CLIMET) covering 243 compounds. We, next, characterize it phenotypically in a diverse panel of myeloid leukemia cell lines and primary patient cells. Analysis of the drug response profiles reveals that 77 drugs affect cell viability, with the top effective compounds targeting nucleic acid synthesis, oxidative stress, and the PI3K/mTOR pathway. Clustering of individual drug response profiles stratifies the cell lines into five functional groups, which link to specific molecular and metabolic features. Mechanistic characterization of selective responses to the PI3K inhibitor pictilisib, the fatty acid synthase inhibitor GSK2194069, and the SLC16A1 inhibitor AZD3965, bring forth biomarkers of drug response. Phenotypic screening using CLIMET represents a valuable tool to probe cellular metabolism and identify metabolic dependencies at large.
Collapse
Affiliation(s)
- Tea Pemovska
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Johannes W Bigenzahn
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Ismet Srndic
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Alexander Lercher
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Andreas Bergthaler
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Adrián César-Razquin
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Felix Kartnig
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Christoph Kornauth
- Department of Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center Vienna, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| | - Peter Valent
- Department of Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Philipp B Staber
- Department of Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center Vienna, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| | - Giulio Superti-Furga
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|