1
|
Shaw S, Jiang W, Rush J, Dumont N, Kim J, Singh R, Skepner A, Khodier C, Raffier C, Murphy Z, Yan N, Schluter C, Yu X, Szuchnicki M, Sathappa M, Kahn J, Sperling AS, Wagner F, McKinney DC, Gould AE, Garvie CW, Miller PG. Identification of small molecule inhibitors of PPM1D using an integrated drug discovery platform. iScience 2025; 28:112069. [PMID: 40124519 PMCID: PMC11930361 DOI: 10.1016/j.isci.2025.112069] [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: 06/25/2024] [Revised: 10/23/2024] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
PPM1D is a serine/threonine phosphatase recurrently activated in cancer, regulates the DNA damage response (DDR), and suppresses p53. Though PPM1D inhibition impairs tumor growth in cancer models and is the subject of multiple drug discovery efforts, no PPM1D inhibitors with clinical potential have been identified. We screened 600,000 compounds in a displacement assay and generated a hit series with nanomolar activity. We optimized our leads using internally developed assays to interrogate PPM1D, p53, and the DDR and defined important structure-activity relationships. Using an in vivo bioluminescent readout of p53 activation, we compared different DDR and p53 modulators and showed that despite having a distinct chemical structure, our lead compound had comparable in vivo activity to established PPM1D inhibitors. Our approach yielded multiple allosteric inhibitors of PPM1D, deepened our understanding of PPM1D as a drug target, and is highly amenable to studying other modulators of the DDR and p53.
Collapse
Affiliation(s)
- Subrata Shaw
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Wei Jiang
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Jason Rush
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Nancy Dumont
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - John Kim
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Ritu Singh
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Adam Skepner
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Carol Khodier
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Cerise Raffier
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Zachary Murphy
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Ni Yan
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Krantz Family Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Cameron Schluter
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Krantz Family Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Xiao Yu
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Mateusz Szuchnicki
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Murugappan Sathappa
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Josephine Kahn
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Adam S. Sperling
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Florence Wagner
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - David C. McKinney
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Alexandra E. Gould
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Colin W. Garvie
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Peter G. Miller
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Krantz Family Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
2
|
Du G, Zheng K, Sun C, Sun M, Pan J, Meng D, Guan W, Zhao H. The relationship mammalian p38 with human health and its homolog Hog1 in response to environmental stresses in Saccharomyces cerevisiae. Front Cell Dev Biol 2025; 13:1522294. [PMID: 40129568 PMCID: PMC11931143 DOI: 10.3389/fcell.2025.1522294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 02/13/2025] [Indexed: 03/26/2025] Open
Abstract
The mammalian p38 MAPK pathway plays a vital role in transducing extracellular environmental stresses into numerous intracellular biological processes. The p38 MAPK have been linked to a variety of cellular processes including inflammation, cell cycle, apoptosis, development and tumorigenesis in specific cell types. The p38 MAPK pathway has been implicated in the development of many human diseases and become a target for treatment of cancer. Although MAPK p38 pathway has been extensively studied, many questions still await clarification. More comprehensive understanding of the MAPK p38 pathway will provide new possibilities for the treatment of human diseases. Hog1 in S. cerevisiae is the conserved homolog of p38 in mammalian cells and the HOG MAPK signaling pathway in S. cerevisiae has been extensively studied. The deep understanding of HOG MAPK signaling pathway will help provide clues for clarifying the p38 signaling pathway, thereby furthering our understanding of the relationship between p38 and disease. In this review, we elaborate the functions of p38 and the relationship between p38 and human disease. while also analyzing how Hog1 regulates cellular processes in response to environmental stresses. 1, p38 in response to various stresses in mammalian cells.2, The functions of mammalian p38 in human health.3, Hog1 as conserved homolog of p38 in response to environmental stresses in Saccharomyces cerevisiae. 1, p38 in response to various stresses in mammalian cells. 2, The functions of mammalian p38 in human health. 3, Hog1 as conserved homolog of p38 in response to environmental stresses in S. cerevisiae.
Collapse
Affiliation(s)
- Gang Du
- *Correspondence: Gang Du, ; Wenqiang Guan, ; Hui Zhao,
| | | | | | | | | | | | - Wenqiang Guan
- Tianjin Key Laboratory of Food Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, China
| | - Hui Zhao
- Tianjin Key Laboratory of Food Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, China
| |
Collapse
|
3
|
Wu K, Ge XX, Duan XF, Li JQ, Wang K, Chen QH, Huang ZM, Zhang WY, Wu Y, Li Q. Wip1 phosphatase activator QGC-8-52 specifically sensitizes p53-negative cancer cells to chemotherapy while protecting normal cells. Drug Resist Updat 2025; 79:101196. [PMID: 39787991 DOI: 10.1016/j.drup.2024.101196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 12/03/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025]
Abstract
PP2C serine-threonine phosphatase Wip1 plays an important role in normal tissue homeostasis, stress signaling and pathogenesis of various human diseases. It is an attractive drug target for cancer treatment and inhibition of its expression or activity constitute a novel therapeutic intervention strategy to prevent the development of various cancers. However, previous strategies for Wip1 suppression may be ineffective in cancers lacking p53. Here, we have characterized the activity of a novel Wip1 phosphatase activator, QGC-8-52, in preclinical models of breast malignancies. QGC-8-52 significantly sensitizes the cancer cell lines with p53 deletion to chemotherapeutic agents. This effect was mediated by the Wip1-FOXO3a interaction and subsequent dephosphorylation of Thr487 that resulted, in response to anticancer treatment, in enhancing the transcription activity of FOXO3a on the proapoptotic TRAIL gene. The sensitizing effect of Wip1 activation on chemotherapeutic drugs only targeted cancer cells lacking p53. The activation of Wip1 in normal cells provided protection from anticancer drug-induced apoptosis by reducing the strength of upstream signaling to p53. Therefore, during the treatment of anticancer drugs, the activated Wip1 phosphatase boosts the apoptosis of p53-negative tumors and protects normal tissues. Our findings may represent an effective and safe therapeutic strategy for cancers with p53 deletion.
Collapse
Affiliation(s)
- Ke Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charls Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA; School of Nursing, Wuhan University, Wuhan, 430071, China
| | - Xiao-Xiao Ge
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Xiao-Fan Duan
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Jie-Qing Li
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Kun Wang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Qiao-Hong Chen
- Department of Chemistry, California State University, Fresno, 2555 E. San Ramon Avenue, M/S SB70, Fresno, CA 93740, USA
| | - Zhi-Min Huang
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | | | - Yong Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charls Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.
| | - Qun Li
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China.
| |
Collapse
|
4
|
Lagorgette L, Bogdanova DA, Belotserkovskaya EV, Garrido C, Demidov ON. PP2C phosphatases-terminators of suicidal thoughts. Cell Death Dis 2024; 15:919. [PMID: 39702569 DOI: 10.1038/s41419-024-07269-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/16/2024] [Accepted: 11/27/2024] [Indexed: 12/21/2024]
Abstract
Cell death and related signaling pathways are essential during development and in various physiological and pathological conditions. Post-translational modifications such as ubiquitination and phosphorylation play an important role in these signaling pathways. The involvement of kinases - enzymes that catalyze protein phosphorylation - in cell death signaling has been extensively studied. On the other hand, not many studies have been devoted to analyzing the role in cell death of phosphatases, enzymes involved in the removal of phosphorylated residues added to proteins by kinases. Obviously, the two opposite reactions, phosphorylation and dephosphorylation, are equally important in the regulation of protein functions and subsequently in the execution of the cell death program. Here, we have summarized recent work on the involvement of serine-threonine PP2C phosphatases in cell death pathways, senescence and autophagy, focusing in particular on the most studied phosphatase PPM1D (PP2Cδ) as an example of the regulatory role of PP2Cs in cell death. The review should help to draw attention to the importance of PP2C family phosphatases in cell death checkpoints and to discover new targets for drug development.
Collapse
Affiliation(s)
- Lisa Lagorgette
- INSERM, UMR 1231, Laboratoire d'Excellence LipSTIC and « Equipe labellisée par la Ligue Nationale contre le Cancer », University of Burgundy, Dijon, France
- University of Burgundy, Faculty of Medicine and Pharmacy, Dijon, France
| | - Daria A Bogdanova
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius University of Science and Technology, Sochi, Russia
- Institute of Cytology RAS, St. Petersburg, Russia
| | | | - Carmen Garrido
- INSERM, UMR 1231, Laboratoire d'Excellence LipSTIC and « Equipe labellisée par la Ligue Nationale contre le Cancer », University of Burgundy, Dijon, France
- University of Burgundy, Faculty of Medicine and Pharmacy, Dijon, France
- Center for Cancer Georges-François Leclerc, Dijon, France
| | - Oleg N Demidov
- INSERM, UMR 1231, Laboratoire d'Excellence LipSTIC and « Equipe labellisée par la Ligue Nationale contre le Cancer », University of Burgundy, Dijon, France.
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius University of Science and Technology, Sochi, Russia.
- Institute of Cytology RAS, St. Petersburg, Russia.
| |
Collapse
|
5
|
Kudo R, Safonov A, Jones C, Moiso E, Dry JR, Shao H, Nag S, da Silva EM, Yildirim SY, Li Q, O'Connell E, Patel P, Will M, Fushimi A, Benitez M, Bradic M, Fan L, Nakshatri H, Sudhan DR, Denz CR, Huerga Sanchez I, Reis-Filho JS, Goel S, Koff A, Weigelt B, Khan QJ, Razavi P, Chandarlapaty S. Long-term breast cancer response to CDK4/6 inhibition defined by TP53-mediated geroconversion. Cancer Cell 2024; 42:1919-1935.e9. [PMID: 39393354 DOI: 10.1016/j.ccell.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 07/02/2024] [Accepted: 09/16/2024] [Indexed: 10/13/2024]
Abstract
Inhibition of CDK4/6 kinases has led to improved outcomes in breast cancer. Nevertheless, only a minority of patients experience long-term disease control. Using a large, clinically annotated cohort of patients with metastatic hormone receptor-positive (HR+) breast cancer, we identify TP53 loss (27.6%) and MDM2 amplification (6.4%) to be associated with lack of long-term disease control. Human breast cancer models reveal that p53 loss does not alter CDK4/6 activity or G1 blockade but instead promotes drug-insensitive p130 phosphorylation by CDK2. The persistence of phospho-p130 prevents DREAM complex assembly, enabling cell-cycle re-entry and tumor progression. Inhibitors of CDK2 can overcome p53 loss, leading to geroconversion and manifestation of senescence phenotypes. Complete inhibition of both CDK4/6 and CDK2 kinases appears to be necessary to facilitate long-term response across genomically diverse HR+ breast cancers.
Collapse
Affiliation(s)
- Rei Kudo
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA; Department of Surgery, The Jikei University School of Medicine, Tokyo 1058461, Japan
| | - Anton Safonov
- Breast Medicine Service, Department of Medicine, MSK, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA
| | - Catherine Jones
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Enrico Moiso
- Department of Medicine, MSK, New York, NY 10065, USA; Department of Epidemiology and Biostatistics, MSK, New York, NY 10065, USA
| | | | - Hong Shao
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Sharanya Nag
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Edaise M da Silva
- Department of Pathology and Laboratory Medicine, MSK, New York, NY 10065, USA
| | - Selma Yeni Yildirim
- Department of Pathology and Laboratory Medicine, MSK, New York, NY 10065, USA
| | - Qing Li
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Elizabeth O'Connell
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Payal Patel
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Marie Will
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA; Breast Medicine Service, Department of Medicine, MSK, New York, NY 10065, USA; Clinical Genetics Service, Department of Medicine, MSK, New York, NY 10065, USA
| | - Atsushi Fushimi
- Department of Surgery, The Jikei University School of Medicine, Tokyo 1058461, Japan
| | - Marimar Benitez
- Program in Molecular Biology, Sloan Kettering Institute, MSK, New York, NY 10065, USA
| | - Martina Bradic
- Program in Molecular Biology, Sloan Kettering Institute, MSK, New York, NY 10065, USA
| | - Li Fan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | - Jorge S Reis-Filho
- Department of Pathology and Laboratory Medicine, MSK, New York, NY 10065, USA
| | - Shom Goel
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Andrew Koff
- Program in Molecular Biology, Sloan Kettering Institute, MSK, New York, NY 10065, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, MSK, New York, NY 10065, USA
| | - Qamar J Khan
- Division of Medical Oncology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Pedram Razavi
- Breast Medicine Service, Department of Medicine, MSK, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA.
| | - Sarat Chandarlapaty
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA; Breast Medicine Service, Department of Medicine, MSK, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
6
|
Pilevneli H, Döger F, Karagenç L, Kozacı D, Kilic Eren M. PPM1D/Wip1 is amplified, overexpressed, and mutated in human non-Hodgkin's lymphomas. Mol Biol Rep 2024; 51:1115. [PMID: 39489796 DOI: 10.1007/s11033-024-10029-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Wip1, is a p53-dependent Ser/Thr phosphatase involved in the timely termination of DDR. The PPM1D gene encoding Wip1 is deregulated and thus gained an oncogene character in common human solid tumors and cell lines. This study assessed the oncogenic potential of the PPM1D gene in human non- Hodgkin's lymphomas (NHL), the most common hematological malignancy worldwide. METHODS AND RESULTS FFPE human lymphoid hyperplasia (LH) (n = 17) and NHL tumor lymph node samples (n = 65) and human NHL cell lines were used to assess the oncogenic potential of the PPM1D gene in the present study. Copy number gain and mRNA expression analysis of the PPM1D/Wip1 gene were assessed by qRT-PCR analysis. Mutational analysis of Exon 6 of the PPM1D gene was performed by PCR amplification and Sanger sequencing. Expressions of Wip1 and p53 proteins were assessed by immunohistochemistry and Western blot analysis. CONCLUSIONS We found that PPM1D gained gene copy number in NHL tumors by 0.7-8 times compared to the control (p < 0.01). Increased PPM1D/Wip1 gene copy number was associated with higher mRNA and protein expression in human NHL samples (p < 0.01). Overexpression of Wip1 in NHL tumors and NHL cell lines was associated with amplification level and was unaffected by p53 status. Furthermore, a heterozygous type insertion mutation was detected in exon 6 (c.1553_1554insA) of the PPM1D gene particularly in DLBCL samples. Wip1 may have oncogenic potential, perhaps playing a role in the onset and progression of human NHL. The possible significance of Wip1 overexpression to chemotherapy response in NHL remains an intriguing question that requires more exploration.
Collapse
Affiliation(s)
- Hatice Pilevneli
- Department of Medical Biology, Faculty of Medicine, Aydın Adnan Menderes University, Aytepe Campus, Aydin, Turkey
| | - Firuzan Döger
- Department of Pathology, Faculty of Medicine, Aydın Adnan Menderes University, Aytepe Campus, Aydin, Turkey
| | - Levent Karagenç
- Department of Histology and Embryology, Faculty of Veterinary Sciences, Aydın Adnan Menderes University, Aydin, Turkey
| | - Didem Kozacı
- Department of Medical Biochemistry, Faculty of Medicine, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Mehtap Kilic Eren
- Department of Medical Biology, Faculty of Medicine, Aydın Adnan Menderes University, Aytepe Campus, Aydin, Turkey.
| |
Collapse
|
7
|
Stoyanov M, Martinikova AS, Matejkova K, Horackova K, Zemankova P, Burdova K, Zemanova Z, Kleiblova P, Kleibl Z, Macurek L. PPM1D activity promotes cellular transformation by preventing senescence and cell death. Oncogene 2024; 43:3081-3093. [PMID: 39237765 PMCID: PMC11473410 DOI: 10.1038/s41388-024-03149-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
Cell cycle checkpoints, oncogene-induced senescence and programmed cell death represent intrinsic barriers to tumorigenesis. Protein phosphatase magnesium-dependent 1 (PPM1D) is a negative regulator of the tumour suppressor p53 and has been implicated in termination of the DNA damage response. Here, we addressed the consequences of increased PPM1D activity resulting from the gain-of-function truncating mutations in exon 6 of the PPM1D. We show that while control cells permanently exit the cell cycle and reside in senescence in the presence of DNA damage caused by ionising radiation or replication stress induced by the active RAS oncogene, RPE1-hTERT and BJ-hTERT cells carrying the truncated PPM1D continue proliferation in the presence of DNA damage, form micronuclei and accumulate genomic rearrangements revealed by karyotyping. Further, we show that increased PPM1D activity promotes cell growth in the soft agar and formation of tumours in xenograft models. Finally, expression profiling of the transformed clones revealed dysregulation of several oncogenic and tumour suppressor pathways. Our data support the oncogenic potential of PPM1D in the context of exposure to ionising radiation and oncogene-induced replication stress.
Collapse
Affiliation(s)
- Miroslav Stoyanov
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Andra S Martinikova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Katerina Matejkova
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Klara Horackova
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Petra Zemankova
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Kamila Burdova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Zuzana Zemanova
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Petra Kleiblova
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Zdenek Kleibl
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Libor Macurek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
8
|
Zhang ZY, Yang ZH, Wang S, Feng SL, Wang XL, Mao JY. Regulation of optimized new Shengmai powder on cardiomyocyte apoptosis and ferroptosis in ischemic heart failure rats: The mediating role of phosphatidylinositol-3-kinase/protein kinase B/tumor protein 53 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118264. [PMID: 38692417 DOI: 10.1016/j.jep.2024.118264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Optimized New Shengmai Powder (ONSMP) is a sophisticated traditional Chinese medicinal formula renowned for bolstering vital energy, optimizing blood circulation, and mitigating fluid retention. After years of clinical application, ONSMP has shown a significant impact in improving myocardial injury and cardiac function and has a positive effect on treating heart failure. However, many unknowns exist about the molecular biological mechanisms of how ONSMP exerts its therapeutic effects, which require further research and exploration. AIM OF THE STUDY Exploring the potential molecular biological mechanisms by which ONSMP ameliorates cardiomyocyte apoptosis and ferroptosis in ischemic heart failure (IHF). MATERIALS AND METHODS First, we constructed a rat model of IHF by inducing acute myocardial infarction through surgery and using echocardiography, organ coefficients, markers of heart failure, antioxidant markers, and histopathological examination to assess the effects of ONSMP on cardiomyocyte apoptosis and ferroptosis in IHF rats. Next, we used bioinformatics analysis techniques to analyze the active components, signaling pathways, and core targets of ONSMP and calculated the interactions between core targets and corresponding elements. Finally, we detected the positive expression of apoptosis and ferroptosis markers and core indicators of signaling pathways by immunohistochemistry; detected the mean fluorescence intensity of core indicators of signaling pathways by immunofluorescence; detected the protein expression of signaling pathways and downstream effector molecules by western blotting; and detected the mRNA levels of p53 and downstream effector molecules by quantitative polymerase chain reaction. RESULTS ONSMP can activate the Ser83 site of ASK by promoting the phosphorylation of the PI3K/AKT axis, thereby inhibiting the MKK3/6-p38 axis and the MKK4/7-JNK axis signaling to reduce p53 expression, and can also directly target and inhibit the activity of p53, ultimately inhibiting p53-mediated mRNA and protein increases in PUMA, SAT1, PIG3, and TFR1, as well as mRNA and protein decreases in SLC7A11, thereby inhibiting cardiomyocyte apoptosis and ferroptosis, effectively improving cardiac function and ventricular remodeling in IHF rat models. CONCLUSION ONSMP can inhibit cardiomyocyte apoptosis and ferroptosis through the PI3K/AKT/p53 signaling pathway, delaying the development of IHF.
Collapse
Affiliation(s)
- Ze-Yu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Zhi-Hua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, PR China.
| | - Shao-Ling Feng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
| | - Xian-Liang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, PR China.
| | - Jing-Yuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, PR China.
| |
Collapse
|
9
|
Kumar JP, Kosek D, Durell SR, Miller Jenkins LM, Debnath S, Coussens NP, Hall MD, Appella DH, Dyda F, Mazur SJ, Appella E. Crystal structure and mechanistic studies of the PPM1D serine/threonine phosphatase catalytic domain. J Biol Chem 2024; 300:107561. [PMID: 39002674 PMCID: PMC11342775 DOI: 10.1016/j.jbc.2024.107561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/15/2024] Open
Abstract
Protein phosphatase 1D (PPM1D, Wip1) is induced by the tumor suppressor p53 during DNA damage response signaling and acts as an oncoprotein in several human cancers. Although PPM1D is a potential therapeutic target, insights into its atomic structure were challenging due to flexible regions unique to this family member. Here, we report the first crystal structure of the PPM1D catalytic domain to 1.8 Å resolution. The structure reveals the active site with two Mg2+ ions bound, similar to other structures. The flap subdomain and B-loop, which are crucial for substrate recognition and catalysis, were also resolved, with the flap forming two short helices and three short β-strands that are followed by an irregular loop. Unexpectedly, a nitrogen-oxygen-sulfur bridge was identified in the catalytic domain. Molecular dynamics simulations and kinetic studies provided further mechanistic insights into the regulation of PPM1D catalytic activity. In particular, the kinetic experiments demonstrated a magnesium concentration-dependent lag in PPM1D attaining steady-state velocity, a feature of hysteretic enzymes that show slow transitions compared with catalytic turnover. All combined, these results advance the understanding of PPM1D function and will support the development of PPM1D-targeted therapeutics.
Collapse
Affiliation(s)
- Jay Prakash Kumar
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Dalibor Kosek
- Laboratory of Molecular Biology, NIDDK, National Institutes of Health, Bethesda, Maryland, United States
| | - Stewart R Durell
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Lisa M Miller Jenkins
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Subrata Debnath
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Nathan P Coussens
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, United States
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, United States
| | - Daniel H Appella
- Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland, United States
| | - Fred Dyda
- Laboratory of Molecular Biology, NIDDK, National Institutes of Health, Bethesda, Maryland, United States
| | - Sharlyn J Mazur
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Ettore Appella
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States.
| |
Collapse
|
10
|
Zhang L, Hsu JI, Braekeleer ED, Chen CW, Patel TD, Martell AG, Guzman AG, Wohlan K, Waldvogel SM, Uryu H, Tovy A, Callen E, Murdaugh RL, Richard R, Jansen S, Vissers L, de Vries BBA, Nussenzweig A, Huang S, Coarfa C, Anastas J, Takahashi K, Vassiliou G, Goodell MA. SOD1 is a synthetic-lethal target in PPM1D-mutant leukemia cells. eLife 2024; 12:RP91611. [PMID: 38896450 PMCID: PMC11186636 DOI: 10.7554/elife.91611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024] Open
Abstract
The DNA damage response is critical for maintaining genome integrity and is commonly disrupted in the development of cancer. PPM1D (protein phosphatase Mg2+/Mn2+-dependent 1D) is a master negative regulator of the response; gain-of-function mutations and amplifications of PPM1D are found across several human cancers making it a relevant pharmacological target. Here, we used CRISPR/Cas9 screening to identify synthetic-lethal dependencies of PPM1D, uncovering superoxide dismutase-1 (SOD1) as a potential target for PPM1D-mutant cells. We revealed a dysregulated redox landscape characterized by elevated levels of reactive oxygen species and a compromised response to oxidative stress in PPM1D-mutant cells. Altogether, our results demonstrate a role for SOD1 in the survival of PPM1D-mutant leukemia cells and highlight a new potential therapeutic strategy against PPM1D-mutant cancers.
Collapse
Affiliation(s)
- Linda Zhang
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of MedicineHoustonUnited States
- Medical Scientist Training Program, Baylor College of MedicineHoustonUnited States
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Center for Cell and Gene TherapyHoustonUnited States
| | - Joanne I Hsu
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of MedicineHoustonUnited States
- Medical Scientist Training Program, Baylor College of MedicineHoustonUnited States
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
| | - Etienne D Braekeleer
- Department of Haematology, Wellcome-MRC Cambridge Stem Cell Institute, University of CambridgeCambridgeUnited Kingdom
| | - Chun-Wei Chen
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Center for Cell and Gene TherapyHoustonUnited States
- Integrated Molecular and Biomedical Sciences Graduate Program, Baylor College of MedicineHoustonUnited States
| | - Tajhal D Patel
- Texas Children’s Hospital Department of Hematology/Oncology, Baylor College of MedicineHoustonUnited States
| | - Alejandra G Martell
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Anna G Guzman
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Katharina Wohlan
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Sarah M Waldvogel
- Medical Scientist Training Program, Baylor College of MedicineHoustonUnited States
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Center for Cell and Gene TherapyHoustonUnited States
- Cancer and Cell Biology Graduate Program, Baylor College of MedicineHoustonUnited States
| | - Hidetaka Uryu
- Department of Leukemia, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | - Ayala Tovy
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Center for Cell and Gene TherapyHoustonUnited States
| | - Elsa Callen
- Laboratory of Genome Integrity, National Cancer Institute, National Institute of HealthBethesdaUnited States
| | - Rebecca L Murdaugh
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Center for Cell and Gene TherapyHoustonUnited States
- Department of Neurosurgery, Baylor College of MedicineHoustonUnited States
| | - Rosemary Richard
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Center for Cell and Gene TherapyHoustonUnited States
- Department of Neurosurgery, Baylor College of MedicineHoustonUnited States
| | - Sandra Jansen
- Donders Centre for Neuroscience, Radboud University Medical CenterNijmegenNetherlands
| | - Lisenka Vissers
- Donders Centre for Neuroscience, Radboud University Medical CenterNijmegenNetherlands
| | - Bert BA de Vries
- Donders Centre for Neuroscience, Radboud University Medical CenterNijmegenNetherlands
| | - Andre Nussenzweig
- Laboratory of Genome Integrity, National Cancer Institute, National Institute of HealthBethesdaUnited States
| | - Shixia Huang
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Department of Education, Innovation and Technology, Advanced Technology Cores, University of TexasHoustonUnited States
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Jamie Anastas
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Center for Cell and Gene TherapyHoustonUnited States
- Department of Neurosurgery, Baylor College of MedicineHoustonUnited States
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer CenterHoustonUnited States
- Department of Genome Medicine, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | - George Vassiliou
- Department of Haematology, Wellcome-MRC Cambridge Stem Cell Institute, University of CambridgeCambridgeUnited Kingdom
| | - Margaret A Goodell
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Center for Cell and Gene TherapyHoustonUnited States
| |
Collapse
|
11
|
Jiang W, Shaw S, Rush J, Dumont N, Kim J, Singh R, Skepner A, Khodier C, Raffier C, Yan N, Schluter C, Yu X, Szuchnicki M, Sathappa M, Kahn J, Sperling AS, McKinney DC, Gould AE, Garvie CW, Miller PG. Identification of Small Molecule Inhibitors of PPM1D Using a Novel Drug Discovery Platform. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.595001. [PMID: 38826457 PMCID: PMC11142126 DOI: 10.1101/2024.05.20.595001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Protein phosphatase, Mg2+/Mn2+ dependent 1D (PPM1D), is a serine/threonine phosphatase that is recurrently activated in cancer, regulates the DNA damage response (DDR), and suppresses the activation of p53. Consistent with its oncogenic properties, genetic loss or pharmacologic inhibition of PPM1D impairs tumor growth and sensitizes cancer cells to cytotoxic therapies in a wide range of preclinical models. Given the therapeutic potential of targeting PPM1D specifically and the DDR and p53 pathway more generally, we sought to deepen our biological understanding of PPM1D as a drug target and determine how PPM1D inhibition differs from other therapeutic approaches to activate the DDR. We performed a high throughput screen to identify new allosteric inhibitors of PPM1D, then generated and optimized a suite of enzymatic, cell-based, and in vivo pharmacokinetic and pharmacodynamic assays to drive medicinal chemistry efforts and to further interrogate the biology of PPM1D. Importantly, this drug discovery platform can be readily adapted to broadly study the DDR and p53. We identified compounds distinct from previously reported allosteric inhibitors and showed in vivo on-target activity. Our data suggest that the biological effects of inhibiting PPM1D are distinct from inhibitors of the MDM2-p53 interaction and standard cytotoxic chemotherapies. These differences also highlight the potential therapeutic contexts in which targeting PPM1D would be most valuable. Therefore, our studies have identified a series of new PPM1D inhibitors, generated a suite of in vitro and in vivo assays that can be broadly used to interrogate the DDR, and provided important new insights into PPM1D as a drug target.
Collapse
Affiliation(s)
- Wei Jiang
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Subrata Shaw
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Jason Rush
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Nancy Dumont
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - John Kim
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Ritu Singh
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Adam Skepner
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Carol Khodier
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Cerise Raffier
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Ni Yan
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Cameron Schluter
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Xiao Yu
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Mateusz Szuchnicki
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Murugappan Sathappa
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Josephine Kahn
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Adam S. Sperling
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David C. McKinney
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Alexandra E. Gould
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Colin W. Garvie
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Peter G. Miller
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
12
|
Gibson CJ, Lindsley RC, Gondek LP. Clonal hematopoiesis in the setting of hematopoietic cell transplantation. Semin Hematol 2024; 61:9-15. [PMID: 38429201 PMCID: PMC10978245 DOI: 10.1053/j.seminhematol.2024.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/12/2024] [Accepted: 01/28/2024] [Indexed: 03/03/2024]
Abstract
Clonal hematopoiesis (CH) in autologous transplant recipients and allogeneic transplant donors has genetic features and clinical associations that are distinct from each other and from non-cancer populations. CH in the setting of autologous transplant is enriched for mutations in DNA damage response pathway genes and is associated with adverse outcomes, including an increased risk of therapy-related myeloid neoplasm and inferior overall survival. Studies of CH in allogeneic transplant donors have yielded conflicting results but have generally shown evidence of potentiated alloimmunity in recipients, with some studies showing an association with favorable recipient outcomes.
Collapse
Affiliation(s)
| | - R Coleman Lindsley
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - Lukasz P Gondek
- Division of Hematologic Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD.
| |
Collapse
|
13
|
Zhang L, Hsu JI, Braekeleer ED, Chen CW, Patel TD, Martell AG, Guzman AG, Wohlan K, Waldvogel SM, Urya H, Tovy A, Callen E, Murdaugh R, Richard R, Jansen S, Vissers L, de Vries BB, Nussenzweig A, Huang S, Coarfa C, Anastas JN, Takahashi K, Vassiliou G, Goodell MA. SOD1 is a synthetic lethal target in PPM1D-mutant leukemia cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.31.555634. [PMID: 37693622 PMCID: PMC10491179 DOI: 10.1101/2023.08.31.555634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The DNA damage response is critical for maintaining genome integrity and is commonly disrupted in the development of cancer. PPM1D (protein phosphatase, Mg2+/Mn2+ dependent 1D) is a master negative regulator of the response; gain-of-function mutations and amplifications of PPM1D are found across several human cancers making it a relevant pharmacologic target. Here, we used CRISPR/Cas9 screening to identify synthetic-lethal dependencies of PPM1D, uncovering superoxide dismutase-1 (SOD1) as a potential target for PPM1D-mutant cells. We revealed a dysregulated redox landscape characterized by elevated levels of reactive oxygen species and a compromised response to oxidative stress in PPM1D-mutant cells. Altogether, our results demonstrate the protective role of SOD1 against oxidative stress in PPM1D-mutant leukemia cells and highlight a new potential therapeutic strategy against PPM1D-mutant cancers.
Collapse
Affiliation(s)
- Linda Zhang
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, TX
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston TX
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Center for Cell and Gene Therapy, Houston, TX
| | - Joanne I. Hsu
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, TX
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston TX
| | - Etienne D. Braekeleer
- Department of Haematology, Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge
| | - Chun-Wei Chen
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston TX
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Center for Cell and Gene Therapy, Houston, TX
- Integrated Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX
| | - Tajhal D. Patel
- Texas Children’s Hospital Department of Hematology/Oncology, Baylor College of Medicine, Houston, TX
| | - Alejandra G. Martell
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Anna G. Guzman
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Katharina Wohlan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Sarah M. Waldvogel
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston TX
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Center for Cell and Gene Therapy, Houston, TX
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, TX
| | - Hidetaka Urya
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ayala Tovy
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston TX
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Center for Cell and Gene Therapy, Houston, TX
| | - Elsa Callen
- Laboratory of Genome Integrity, National Cancer Institute, National Institute of Health, Bethesda, MD
| | - Rebecca Murdaugh
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston TX
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Center for Cell and Gene Therapy, Houston, TX
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX
| | - Rosemary Richard
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston TX
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Center for Cell and Gene Therapy, Houston, TX
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX
| | - Sandra Jansen
- Donders Centre for Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lisenka Vissers
- Donders Centre for Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bert B.A. de Vries
- Donders Centre for Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Andre Nussenzweig
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, TX
| | - Shixia Huang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Department of Education, Innovation and Technology, Advanced Technology Cores
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Jamie N. Anastas
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston TX
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Center for Cell and Gene Therapy, Houston, TX
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Education, Innovation and Technology, Advanced Technology Cores
| | - George Vassiliou
- Department of Haematology, Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge
| | - Margaret A. Goodell
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston TX
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Center for Cell and Gene Therapy, Houston, TX
| |
Collapse
|
14
|
Martinikova AS, Stoyanov M, Oravetzova A, Kok YP, Yu S, Dobrovolna J, Janscak P, van Vugt M, Macurek L. PPM1D activity promotes the replication stress caused by cyclin E1 overexpression. Mol Oncol 2024; 18:6-20. [PMID: 37067201 PMCID: PMC10766204 DOI: 10.1002/1878-0261.13433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/09/2023] [Accepted: 04/13/2023] [Indexed: 04/18/2023] Open
Abstract
Oncogene-induced replication stress has been recognized as a major cause of genome instability in cancer cells. Increased expression of cyclin E1 caused by amplification of the CCNE1 gene is a common cause of replication stress in various cancers. Protein phosphatase magnesium-dependent 1 delta (PPM1D) is a negative regulator of p53 and has been implicated in termination of the cell cycle checkpoint. Amplification of the PPM1D gene or frameshift mutations in its final exon promote tumorigenesis. Here, we show that PPM1D activity further increases the replication stress caused by overexpression of cyclin E1. In particular, we demonstrate that cells expressing a truncated mutant of PPM1D progress faster from G1 to S phase and fail to complete licensing of the replication origins. In addition, we show that transcription-replication collisions and replication fork slowing caused by CCNE1 overexpression are exaggerated in cells expressing the truncated PPM1D. Finally, replication speed and accumulation of focal DNA copy number alterations caused by induction of CCNE1 expression was rescued by pharmacological inhibition of PPM1D. We propose that increased activity of PPM1D suppresses the checkpoint function of p53 and thus promotes genome instability in cells expressing the CCNE1 oncogene.
Collapse
Affiliation(s)
- Andra S. Martinikova
- Laboratory of Cancer Cell Biology, Institute of Molecular GeneticsCzech Academy of SciencesPragueCzech Republic
| | - Miroslav Stoyanov
- Laboratory of Cancer Cell Biology, Institute of Molecular GeneticsCzech Academy of SciencesPragueCzech Republic
| | - Anna Oravetzova
- Laboratory of Cancer Cell Biology, Institute of Molecular GeneticsCzech Academy of SciencesPragueCzech Republic
| | - Yannick P. Kok
- Department of Medical Oncology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Shibo Yu
- Department of Pathology and Medical Biology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Jana Dobrovolna
- Laboratory of Cancer Cell Biology, Institute of Molecular GeneticsCzech Academy of SciencesPragueCzech Republic
| | - Pavel Janscak
- Laboratory of Cancer Cell Biology, Institute of Molecular GeneticsCzech Academy of SciencesPragueCzech Republic
- Institute of Molecular Cancer ResearchUniversity of ZurichSwitzerland
| | - Marcel van Vugt
- Department of Medical Oncology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Libor Macurek
- Laboratory of Cancer Cell Biology, Institute of Molecular GeneticsCzech Academy of SciencesPragueCzech Republic
| |
Collapse
|
15
|
Seipel K, Frey M, Nilius H, Akhoundova D, Banz Y, Bacher U, Pabst T. Low-Frequency PPM1D Gene Mutations Affect Treatment Response to CD19-Targeted CAR T-Cell Therapy in Large B-Cell Lymphoma. Curr Oncol 2023; 30:10463-10476. [PMID: 38132396 PMCID: PMC10742331 DOI: 10.3390/curroncol30120762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/20/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Chimeric antigen receptor T (CAR T)-cell therapy has become a standard treatment option for patients with relapsed or refractory diffuse large B-cell lymphoma (r/r DLBCL). Mutations in the PPM1D gene, a frequent driver alteration in clonal hematopoiesis (CH), lead to a gain of function of PPM1D/Wip1 phosphatase, impairing p53-dependent G1 checkpoint and promoting cell proliferation. The presence of PPM1D mutations has been correlated with reduced response to standard chemotherapy in lymphoma patients. In this study, we analyzed the impact of low-frequency PPM1D mutations on the safety and efficacy of CD19-targeted CAR T-cell therapy in a cohort of 85 r/r DLBCL patients. In this cohort, the prevalence of PPM1D gene mutations was 20% with a mean variant allele frequency (VAF) of 0.052 and a median VAF of 0.036. CAR T-induced cytokine release syndrome (CRS) and immune effector cell-associated neuro-toxicities (ICANS) occurred at similar frequencies in patients with and without PPM1D mutations. Clinical outcomes were globally worse in the PPM1D mutated (PPM1Dmut) vs. PPM1D wild type (PPM1Dwt) subset. While the prevalent treatment outcome within the PPM1Dwt subgroup was complete remission (56%), the majority of patients within the PPM1Dmut subgroup had only partial remission (60%). Median progression-free survival (PFS) was 3 vs. 12 months (p = 0.07) and median overall survival (OS) was 5 vs. 37 months (p = 0.004) for the PPM1Dmut and PPM1Dwt cohort, respectively. Our data suggest that the occurrence of PPM1D mutations in the context of CH may predict worse outcomes after CD19-targeted CAR T-cell therapy in patients with r/r DLBCL.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/adverse effects
- Receptors, Chimeric Antigen
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/therapeutic use
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Treatment Outcome
- Antigens, CD19/genetics
- Antigens, CD19/therapeutic use
- Protein Phosphatase 2C/genetics
Collapse
Affiliation(s)
- Katja Seipel
- Department for Biomedical Research (DBMR), University of Bern, 3008 Bern, Switzerland;
- Department of Medical Oncology, University Hospital Bern, 3010 Bern, Switzerland;
| | - Michèle Frey
- Department of Medical Oncology, University Hospital Bern, 3010 Bern, Switzerland;
| | - Henning Nilius
- Department of Clinical Chemistry, University of Bern, 3010 Bern, Switzerland;
| | - Dilara Akhoundova
- Department for Biomedical Research (DBMR), University of Bern, 3008 Bern, Switzerland;
- Department of Medical Oncology, University Hospital Bern, 3010 Bern, Switzerland;
| | - Yara Banz
- Institute of Tissue Medicine and Pathology (IGMP), University of Bern, 3010 Bern, Switzerland;
| | - Ulrike Bacher
- Department of Hematology, University Hospital Bern, 3010 Bern, Switzerland;
| | - Thomas Pabst
- Department of Medical Oncology, University Hospital Bern, 3010 Bern, Switzerland;
| |
Collapse
|
16
|
Burocziova M, Danek P, Oravetzova A, Chalupova Z, Alberich-Jorda M, Macurek L. Ppm1d truncating mutations promote the development of genotoxic stress-induced AML. Leukemia 2023; 37:2209-2220. [PMID: 37709843 PMCID: PMC10624630 DOI: 10.1038/s41375-023-02030-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 09/16/2023]
Abstract
Hematopoietic stem cells (HSCs) ensure blood cell production during the life-time of an organism, and to do so they need to balance self-renewal, proliferation, differentiation, and migration in a steady state as well as in response to stress or injury. Importantly, aberrant proliferation of HSCs leads to hematological malignancies, and thus, tight regulation by various tumor suppressor pathways, including p53, is essential. Protein phosphatase magnesium-dependent 1 delta (PPM1D) is a negative regulator of p53 and promotes cell survival upon induction of genotoxic stress. Truncating mutations in the last exon of PPM1D lead to the production of a stable, enzymatically active protein and are commonly associated with clonal hematopoiesis. Using a transgenic mouse model, we demonstrate that truncated PPM1D reduces self-renewal of HSCs in basal conditions but promotes the development of aggressive AML after exposure to ionizing radiation. Inhibition of PPM1D suppressed the colony growth of leukemic stem and progenitor cells carrying the truncated PPM1D, and remarkably, it provided protection against irradiation-induced cell growth. Altogether, we demonstrate that truncated PPM1D affects HSC maintenance, disrupts normal hematopoiesis, and that its inhibition could be beneficial in the context of therapy-induced AML.
Collapse
Affiliation(s)
- Monika Burocziova
- Department Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Prague, Czech Republic
- Department of Hemato-oncology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Prague, Czech Republic
| | - Petr Danek
- Department of Hemato-oncology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Prague, Czech Republic
| | - Anna Oravetzova
- Department Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Prague, Czech Republic
| | - Zuzana Chalupova
- Department Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Prague, Czech Republic
| | - Meritxell Alberich-Jorda
- Department of Hemato-oncology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Prague, Czech Republic.
- Childhood Leukaemia Investigation Prague, Department of Pediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague, University Hospital Motol, V Uvalu 84, Praha, 150 06, Czech Republic.
| | - Libor Macurek
- Department Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Prague, Czech Republic.
| |
Collapse
|
17
|
Belotserkovskaya E, Golotin V, Uyanik B, Demidov ON. Clonal haematopoiesis - a novel entity that modifies pathological processes in elderly. Cell Death Discov 2023; 9:345. [PMID: 37726289 PMCID: PMC10509183 DOI: 10.1038/s41420-023-01590-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 06/02/2023] [Accepted: 07/31/2023] [Indexed: 09/21/2023] Open
Abstract
Progress in the development of new sequencing techniques with wider accessibility and higher sensitivity of the protocol of deciphering genome particularities led to the discovery of a new phenomenon - clonal haematopoiesis. It is characterized by the presence in the bloodstream of elderly people a minor clonal population of cells with mutations in certain genes, but without any sign of disease related to the hematopoietic system. Here we will review this recent advancement in the field of clonal haematopoiesis and how it may affect the disease's development in old age.
Collapse
Affiliation(s)
| | - Vasily Golotin
- Institute of Cytology RAS, 4 Tikhoretskii prospect, St. Petersburg, 194064, Russia
- Saint Petersburg bra-nch of "VNIRO" ("Gos-NOIRH" named after L.S. Berg), Saint Petersburg, Russia
| | - Burhan Uyanik
- INSERM UMR1231, Laboratory of Excellence LipSTIC and label Ligue Nationale contre le Cancer, 7 Boulevard Jeanne d'Arc, Dijon, 21000, France
| | - Oleg N Demidov
- Institute of Cytology RAS, 4 Tikhoretskii prospect, St. Petersburg, 194064, Russia.
- INSERM UMR1231, Laboratory of Excellence LipSTIC and label Ligue Nationale contre le Cancer, 7 Boulevard Jeanne d'Arc, Dijon, 21000, France.
- Sirius University of Science and Technology, 1 Olimpiiskii pr-t, Sochi, 354340, Russian Federation.
| |
Collapse
|
18
|
Attiq A, Afzal S. Trinity of inflammation, innate immune cells and cross-talk of signalling pathways in tumour microenvironment. Front Pharmacol 2023; 14:1255727. [PMID: 37680708 PMCID: PMC10482416 DOI: 10.3389/fphar.2023.1255727] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 08/08/2023] [Indexed: 09/09/2023] Open
Abstract
Unresolved inflammation is a pathological consequence of persistent inflammatory stimulus and perturbation in regulatory mechanisms. It increases the risk of tumour development and orchestrates all stages of tumorigenesis in selected organs. In certain cancers, inflammatory processes create the appropriate conditions for neoplastic transformation. While in other types, oncogenic changes pave the way for an inflammatory microenvironment that leads to tumour development. Of interest, hallmarks of tumour-promoting and cancer-associated inflammation are striking similar, sharing a complex network of stromal (fibroblasts and vascular cells) and inflammatory immune cells that collectively form the tumour microenvironment (TME). The cross-talks of signalling pathways initially developed to support homeostasis, change their role, and promote atypical proliferation, survival, angiogenesis, and subversion of adaptive immunity in TME. These transcriptional and regulatory pathways invariably contribute to cancer-promoting inflammation in chronic inflammatory disorders and foster "smouldering" inflammation in the microenvironment of various tumour types. Besides identifying common target sites of numerous cancer types, signalling programs and their cross-talks governing immune cells' plasticity and functional diversity can be used to develop new fate-mapping and lineage-tracing mechanisms. Here, we review the vital molecular mechanisms and pathways that establish the connection between inflammation and tumour development, progression, and metastasis. We also discussed the cross-talks between signalling pathways and devised strategies focusing on these interaction mechanisms to harness synthetic lethal drug combinations for targeted cancer therapy.
Collapse
Affiliation(s)
- Ali Attiq
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Sheryar Afzal
- Department of Biomedical Sciences, Faculty of Veterinary Medicine, King Faisal University, Al Ahsa, Saudi Arabia
| |
Collapse
|
19
|
Fito-Lopez B, Salvadores M, Alvarez MM, Supek F. Prevalence, causes and impact of TP53-loss phenocopying events in human tumors. BMC Biol 2023; 21:92. [PMID: 37095494 PMCID: PMC10127307 DOI: 10.1186/s12915-023-01595-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/12/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND TP53 is a master tumor suppressor gene, mutated in approximately half of all human cancers. Given the many regulatory roles of the corresponding p53 protein, it is possible to infer loss of p53 activity - which may occur due to alterations in trans - from gene expression patterns. Several such alterations that phenocopy p53 loss are known, however additional ones may exist, but their identity and prevalence among human tumors are not well characterized. RESULTS We perform a large-scale statistical analysis on transcriptomes of ~ 7,000 tumors and ~ 1,000 cell lines, estimating that 12% and 8% of tumors and cancer cell lines, respectively, phenocopy TP53 loss: they are likely deficient in the activity of the p53 pathway, while not bearing obvious TP53 inactivating mutations. While some of these cases are explained by amplifications in the known phenocopying genes MDM2, MDM4 and PPM1D, many are not. An association analysis of cancer genomic scores jointly with CRISPR/RNAi genetic screening data identified an additional common TP53-loss phenocopying gene, USP28. Deletions in USP28 are associated with a TP53 functional impairment in 2.9-7.6% of breast, bladder, lung, liver and stomach tumors, and have comparable effect size to MDM4 amplifications. Additionally, in the known copy number alteration (CNA) segment harboring MDM2, we identify an additional co-amplified gene (CNOT2) that may cooperatively boost the TP53 functional inactivation effect of MDM2. An analysis of cancer cell line drug screens using phenocopy scores suggests that TP53 (in)activity commonly modulates associations between anticancer drug effects and various genetic markers, such as PIK3CA and PTEN mutations, and should thus be considered as a drug activity modifying factor in precision medicine. As a resource, we provide the drug-genetic marker associations that differ depending on TP53 functional status. CONCLUSIONS Human tumors that do not bear obvious TP53 genetic alterations but that phenocopy p53 activity loss are common, and the USP28 gene deletions are one likely cause.
Collapse
Affiliation(s)
- Bruno Fito-Lopez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Marina Salvadores
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Miguel-Martin Alvarez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Fran Supek
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
| |
Collapse
|
20
|
Storchova R, Palek M, Palkova N, Veverka P, Brom T, Hofr C, Macurek L. Phosphorylation of TRF2 promotes its interaction with TIN2 and regulates DNA damage response at telomeres. Nucleic Acids Res 2023; 51:1154-1172. [PMID: 36651296 PMCID: PMC9943673 DOI: 10.1093/nar/gkac1269] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/25/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Protein phosphatase magnesium-dependent 1 delta (PPM1D) terminates the cell cycle checkpoint by dephosphorylating the tumour suppressor protein p53. By targeting additional substrates at chromatin, PPM1D contributes to the control of DNA damage response and DNA repair. Using proximity biotinylation followed by proteomic analysis, we identified a novel interaction between PPM1D and the shelterin complex that protects telomeric DNA. In addition, confocal microscopy revealed that endogenous PPM1D localises at telomeres. Further, we found that ATR phosphorylated TRF2 at S410 after induction of DNA double strand breaks at telomeres and this modification increased after inhibition or loss of PPM1D. TRF2 phosphorylation stimulated its interaction with TIN2 both in vitro and at telomeres. Conversely, induced expression of PPM1D impaired localisation of TIN2 and TPP1 at telomeres. Finally, recruitment of the DNA repair factor 53BP1 to the telomeric breaks was strongly reduced after inhibition of PPM1D and was rescued by the expression of TRF2-S410A mutant. Our results suggest that TRF2 phosphorylation promotes the association of TIN2 within the shelterin complex and regulates DNA repair at telomeres.
Collapse
Affiliation(s)
- Radka Storchova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague CZ-14220, Czech Republic
| | - Matous Palek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague CZ-14220, Czech Republic
| | - Natalie Palkova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague CZ-14220, Czech Republic
| | - Pavel Veverka
- LifeB, Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno CZ-62500, Czech Republic
| | - Tomas Brom
- LifeB, Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno CZ-62500, Czech Republic
| | - Ctirad Hofr
- LifeB, Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno CZ-62500, Czech Republic
| | - Libor Macurek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague CZ-14220, Czech Republic
| |
Collapse
|
21
|
Liu Y, Gu S, Su Y, Wang S, Cheng Y, Sang X, Jin L, Liu Y, Li C, Liu W, Chen M, Wang X, Wang Z. Embryonic stem cell extracellular vesicles reverse the senescence of retinal pigment epithelial cells by the p38MAPK pathway. Exp Eye Res 2023; 227:109365. [PMID: 36577484 DOI: 10.1016/j.exer.2022.109365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/26/2022]
Abstract
Retinal pigment epithelial (RPE) cellular senescence is regarded as an initiator for age-related macular degeneration (AMD). We previously demonstrated that by the coculture way, embryonic stem cells (ESCs) can reverse the senescence of RPE cells, but xenograft cells can cause a plethora of adverse effects. Extracellular vesicles (EVs) derived from ESCs can act as messengers to mediate nearby cell activities and have the same potential as ESCs to reverse RPE senescence. Furthermore, ESC-EVs have achieved preliminary efficacy while treating many age-related diseases. The present study aimed to test the effect of ESC-EVs on the replicative senescence model of RPE cells as well as its mechanism. The results showed that ESC-EVs enhanced the proliferative ability and cell cycle transition of senescent RPE cells, whereas reduced the senescence-associated galactosidase (SA-β-gal) staining rate, as well as the levels of mitochondrial membrane potential (MMP) and reactive oxygen species (ROS). Moreover, classical markers of cellular senescence p21WAF1/CIP1 (p21) and p16INK4a (p16) were downregulated. The bioinformatic analysis and further study showed that the inhibition of the p38MAPK pathway by ESC-EVs played a pivotal role in RPE cellular senescence-reversing effect, which was ameliorated or even abolished when dehydrocorydaline were administrated simultaneously, demonstrating that ESC-EVs can effectively reverse RPE cellular senesence by inhibiting the p38MAPK pathway, thus highlights the potential of ESC-derived EVs as biomaterials for preventative and protective therapy in AMD.
Collapse
Affiliation(s)
- Yurun Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| | - Simin Gu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| | - Yaru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| | - Shoubi Wang
- The First Affiliated Hospital of Xiamen, 55 Zhenhai Road, Xiamen, China.
| | - Yaqi Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| | - Xuan Sang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| | - Lin Jin
- The First Affiliated Hospital of Shandong First Medical University, 16766 Jingshi Road, Jinan, Shandong Province, China.
| | - Ying Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| | - Chaoyang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| | - Weiqin Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| | - Minghao Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| | - Xiaoran Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| | - Zhichong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| |
Collapse
|
22
|
Robello M, Zheng H, Saha M, George Rosenker KM, Debnath S, Kumar JP, Tagad HD, Mazur SJ, Appella E, Appella DH. Alkyl-substituted N-methylaryl-N'-aryl-4-aminobenzamides: A new series of small molecule inhibitors for Wip1 phosphatase. Eur J Med Chem 2022; 243:114763. [PMID: 36179402 PMCID: PMC9664485 DOI: 10.1016/j.ejmech.2022.114763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/26/2022] [Accepted: 09/07/2022] [Indexed: 11/03/2022]
Abstract
The wild-type p53 induced phosphatase 1 (Wip1), a member of the serine/threonine-specific PP2C family, is overexpressed in numerous human cancers. Wip1 dephosphorylates p53 as well as several kinases (such as p38 MAPK, ATM, Chk1, and Chk2) in the DNA damage response pathway that are responsible for maintaining genomic stability and preventing oncogenic transformation. As a result, Wip1 is an attractive target for synthetic inhibitors that could be further developed into therapeutics to treat some cancers. In this study, we report a series of alkyl-substituted N-methylaryl-N'-aryl-4-aminobenzamides and their inhibitory activity of the Wip1 phosphatase. A straightforward synthetic route was developed to synthesize the target compounds from commercially available starting materials. Three different portions (R1, R2, R3) of the core scaffold were extensively modified to examine structure-activity relationships. This study revealed interesting trends about a new molecular scaffold to inhibit Wip1.
Collapse
Affiliation(s)
- Marco Robello
- Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, United States
| | - Hongchao Zheng
- Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, United States
| | - Mrinmoy Saha
- Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, United States
| | - Kara M George Rosenker
- Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, United States
| | - Subrata Debnath
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Jay Prakash Kumar
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Harichandra D Tagad
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Sharlyn J Mazur
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Ettore Appella
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Daniel H Appella
- Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, United States.
| |
Collapse
|
23
|
Clausse V, Fang Y, Tao D, Tagad HD, Sun H, Wang Y, Karavadhi S, Lane K, Shi ZD, Vasalatiy O, LeClair CA, Eells R, Shen M, Patnaik S, Appella E, Coussens NP, Hall MD, Appella DH. Discovery of Novel Small-Molecule Scaffolds for the Inhibition and Activation of WIP1 Phosphatase from a RapidFire Mass Spectrometry High-Throughput Screen. ACS Pharmacol Transl Sci 2022; 5:993-1006. [PMID: 36268125 PMCID: PMC9578142 DOI: 10.1021/acsptsci.2c00147] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Indexed: 11/28/2022]
Abstract
Wild-type P53-induced phosphatase 1 (WIP1), also known as PPM1D or PP2Cδ, is a serine/threonine protein phosphatase induced by P53 after genotoxic stress. WIP1 inhibition has been proposed as a therapeutic strategy for P53 wild-type cancers in which it is overexpressed, but this approach would be ineffective in P53-negative cancers. Furthermore, there are several cancers with mutated P53 where WIP1 acts as a tumor suppressor. Therefore, activating WIP1 phosphatase might also be a therapeutic strategy, depending on the P53 status. To date, no specific, potent WIP1 inhibitors with appropriate pharmacokinetic properties have been reported, nor have WIP1-specific activators. Here, we report the discovery of new WIP1 modulators from a high-throughput screen (HTS) using previously described orthogonal biochemical assays suitable for identifying both inhibitors and activators. The primary HTS was performed against a library of 102 277 compounds at a single concentration using a RapidFire mass spectrometry assay. Hits were further evaluated over a range of 11 concentrations with both the RapidFire MS assay and an orthogonal fluorescence-based assay. Further biophysical, biochemical, and cell-based studies of confirmed hits revealed a WIP1 activator and two inhibitors, one competitive and one uncompetitive. These new scaffolds are prime candidates for optimization which might enable inhibitors with improved pharmacokinetics and a first-in-class WIP1 activator.
Collapse
Affiliation(s)
- Victor Clausse
- Synthetic
Bioactive Molecules Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Yuhong Fang
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Dingyin Tao
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Harichandra D. Tagad
- Laboratory
of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Hongmao Sun
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Yuhong Wang
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Surendra Karavadhi
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Kelly Lane
- Chemistry
and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Zhen-Dan Shi
- Chemistry
and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Olga Vasalatiy
- Chemistry
and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Christopher A. LeClair
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Rebecca Eells
- Reaction
Biology Corporation, 1 Great Valley Parkway, Suite 2, Malvern, Pennsylvania 19355, United States
| | - Min Shen
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Samarjit Patnaik
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Ettore Appella
- Laboratory
of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Nathan P. Coussens
- Molecular
Pharmacology Laboratories, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, United States
| | - Matthew D. Hall
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Daniel H. Appella
- Synthetic
Bioactive Molecules Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
24
|
Zhang L, Hsu JI, Goodell MA. PPM1D in Solid and Hematologic Malignancies: Friend and Foe? Mol Cancer Res 2022; 20:1365-1378. [PMID: 35657598 PMCID: PMC9437564 DOI: 10.1158/1541-7786.mcr-21-1018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/26/2022] [Accepted: 06/01/2022] [Indexed: 01/07/2023]
Abstract
In the face of constant genomic insults, the DNA damage response (DDR) is initiated to preserve genome integrity; its disruption is a classic hallmark of cancer. Protein phosphatase Mg2+/Mn2+-dependent 1D (PPM1D) is a central negative regulator of the DDR that is mutated or amplified in many solid cancers. PPM1D overexpression is associated with increased proliferative and metastatic behavior in multiple solid tumor types and patients with PPM1D-mutated malignancies have poorer prognoses. Recent findings have sparked an interest in the role of PPM1D in hematologic malignancies. Acquired somatic mutations may provide hematopoietic stem cells with a competitive advantage, leading to a substantial proportion of mutant progeny in the peripheral blood, an age-associated phenomenon termed "clonal hematopoiesis" (CH). Recent large-scale genomic studies have identified PPM1D to be among the most frequently mutated genes found in individuals with CH. While PPM1D mutations are particularly enriched in patients with therapy-related myeloid neoplasms, their role in driving leukemic transformation remains uncertain. Here, we examine the mechanisms through which PPM1D overexpression or mutation may drive malignancy by suppression of DNA repair, cell-cycle arrest, and apoptosis. We also discuss the divergent roles of PPM1D in the oncogenesis of solid versus hematologic cancers with a view to clinical implications and new therapeutic avenues.
Collapse
Affiliation(s)
- Linda Zhang
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, Texas
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas
| | - Joanne I. Hsu
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, Texas
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas
| | - Margaret A. Goodell
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Corresponding Author: Margaret A. Goodell, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030. E-mail:
| |
Collapse
|
25
|
Yu H, Zou D, Ni N, Zhang S, Zhang Q, Yang L. Overexpression of NCAPG in ovarian cancer is associated with ovarian cancer proliferation and apoptosis via p38 MAPK signaling pathway. J Ovarian Res 2022; 15:98. [PMID: 35986371 PMCID: PMC9389752 DOI: 10.1186/s13048-022-01030-z] [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] [Received: 07/05/2021] [Accepted: 08/10/2022] [Indexed: 12/24/2022] Open
Abstract
Background Non-SMC condensin I complex subunit G (NCAPG), a member of the subunit of condensin complex, is significantly overexpressed in various cancers and involved in the pathogenesis of cancers. However, the roles of NCAPG in ovarian cancer remain unclear. Methods The mRNA expression, overall survival, and disease-free survival of NCAPG in ovarian cancer were analyzed by GEPIA and KM plotter database, and the expression levels of NCAPG in OC tissues and cell lines were determined by qPCR and immunohistochemistry analysis. shRNA targeting NCAPG gene (sh-NCAPG) was utilized to knock down NCAPG expression in OVCAR3 and SKOV3 cells. Subsequently, CCK-8 assay, colony formation assay, transwell invasion assay and flow cytometric analysis were performed to detect the effect of NCAPG on OC cell proliferation, apoptosis, and invasion. Finally, western blot assays were performed to detect the mechanism of NCAPG in ovarian cancer. Results Analysis using GEPIA and KM plotter database showed NCAPG was upregulated in ovarian cancer and negatively associated with the survival of OC patients. qPCR and immunohistochemistry analysis confirmed it was highly expressed in both ovarian cancer tissues and cells. The silencing of NCAPG inhibited OC cell proliferation and invasion, and induced cell apoptosis. Additionally, flow cytometric analysis revealed that NCAPG knockdown arrested the cell cycle at G2 and S phases. Furthermore, we also found that downregulation of NCAPG could suppress OC cell proliferation and invasion via activating the p38 MAPK signaling pathway. Conclusion Our results suggest that NCAPG exhibits an important role in the development and progression of ovarian cancer and implicates NCAPG as a potential therapeutic target in ovarian cancer.
Collapse
|
26
|
Miller PG, Sathappa M, Moroco JA, Jiang W, Qian Y, Iqbal S, Guo Q, Giacomelli AO, Shaw S, Vernier C, Bajrami B, Yang X, Raffier C, Sperling AS, Gibson CJ, Kahn J, Jin C, Ranaghan M, Caliman A, Brousseau M, Fischer ES, Lintner R, Piccioni F, Campbell AJ, Root DE, Garvie CW, Ebert BL. Allosteric inhibition of PPM1D serine/threonine phosphatase via an altered conformational state. Nat Commun 2022; 13:3778. [PMID: 35773251 PMCID: PMC9246869 DOI: 10.1038/s41467-022-30463-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 05/02/2022] [Indexed: 02/02/2023] Open
Abstract
PPM1D encodes a serine/threonine phosphatase that regulates numerous pathways including the DNA damage response and p53. Activating mutations and amplification of PPM1D are found across numerous cancer types. GSK2830371 is a potent and selective allosteric inhibitor of PPM1D, but its mechanism of binding and inhibition of catalytic activity are unknown. Here we use computational, biochemical and functional genetic studies to elucidate the molecular basis of GSK2830371 activity. These data confirm that GSK2830371 binds an allosteric site of PPM1D with high affinity. By further incorporating data from hydrogen deuterium exchange mass spectrometry and sedimentation velocity analytical ultracentrifugation, we demonstrate that PPM1D exists in an equilibrium between two conformations that are defined by the movement of the flap domain, which is required for substrate recognition. A hinge region was identified that is critical for switching between the two conformations and was directly implicated in the high-affinity binding of GSK2830371 to PPM1D. We propose that the two conformations represent active and inactive forms of the protein reflected by the position of the flap, and that binding of GSK2830371 shifts the equilibrium to the inactive form. Finally, we found that C-terminal truncating mutations proximal to residue 400 result in destabilization of the protein via loss of a stabilizing N- and C-terminal interaction, consistent with the observation from human genetic data that nearly all PPM1D mutations in cancer are truncating and occur distal to residue 400. Taken together, our findings elucidate the mechanism by which binding of a small molecule to an allosteric site of PPM1D inhibits its activity and provides insights into the biology of PPM1D.
Collapse
Affiliation(s)
- Peter G Miller
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Murugappan Sathappa
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Jamie A Moroco
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Wei Jiang
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Yue Qian
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Sumaiya Iqbal
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Qi Guo
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Andrew O Giacomelli
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Subrata Shaw
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Camille Vernier
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Besnik Bajrami
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Xiaoping Yang
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Cerise Raffier
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Adam S Sperling
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Christopher J Gibson
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Josephine Kahn
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Cyrus Jin
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Matthew Ranaghan
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Alisha Caliman
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Merissa Brousseau
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Eric S Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Robert Lintner
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | | | | | - David E Root
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Colin W Garvie
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA.
| | - Benjamin L Ebert
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Bethesda, MD, USA.
| |
Collapse
|
27
|
Yin S, Yang L, Zheng Y, Zang R. MS: Wip1 suppresses angiogenesis through the STAT3-VEGF signalling pathway in serous ovarian cancer. J Ovarian Res 2022; 15:56. [PMID: 35538489 PMCID: PMC9087943 DOI: 10.1186/s13048-022-00990-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/25/2022] [Indexed: 12/20/2022] Open
Abstract
Multifaceted functions of the so-called “oncogene” Wip1 have been reported in a previous study, while its actual role remains to be explored in serous ovarian cancer (SOC). In this study, by performing bioinformatic analysis with a public database and immunohistochemical staining of Wip1 in tumour tissue from SOC, we concluded that decreased expression of Wip1 was associated with a higher rate of tumour metastasis and platinum-based therapy resistance and increased ascites volume, which led to poorer prognosis in SOC patients. We also found that overexpression of Wip1 in SKOV3 cells decreased the levels of several cytokines, including VEGF, by secretome profiling analysis, and Wip1 overexpression suppressed angiogenesis both in vitro and in vivo. Mechanistic studies indicated that overexpression of Wip1 decreased the expression of VEGF at both the protein and mRNA levels and that the inhibitory effect was mediated by dephosphorylation of STAT3 at Ser727. Our study uncovered the role of Wip1 in SOC and provides a novel therapeutic strategy for suppressing angiogenesis.
Collapse
Affiliation(s)
- Sheng Yin
- Department of Gynaecologic Oncology, Ovarian Cancer Program, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lina Yang
- Department of Obstetrics and Gynecology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yiyan Zheng
- Department of Gynaecologic Oncology, Ovarian Cancer Program, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rongyu Zang
- Department of Gynaecologic Oncology, Ovarian Cancer Program, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
28
|
Davey MG, Feeney G, Annuk H, Paganga M, Holian E, Lowery AJ, Kerin MJ, Miller N. MicroRNA Expression Profiling Predicts Nodal Status and Disease Recurrence in Patients Treated with Curative Intent for Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14092109. [PMID: 35565239 PMCID: PMC9106021 DOI: 10.3390/cancers14092109] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/13/2022] [Accepted: 04/20/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Approximately one-third of colorectal cancer (CRC) patients will suffer recurrence. MiRNAs are small non-coding RNAs that play important roles in gene expression. We aimed to correlate miRNA expression with aggressive clinicopathological characteristics and survival outcomes in CRC. Methods: Tumour samples were extracted from 74 CRC patients. MiRNAs were quantified using real-time reverse transcriptase polymerase chain reaction. Descriptive statistics and Cox regression analyses were performed to correlate miRNA targets with clinicopathological and outcome data. Results: Aberrant miR-21 and miR-135b expression correlate with increased nodal stage (p = 0.039, p = 0.022). Using univariable Cox regression analyses, reduced miR-135b (β-coefficient −1.126, hazard ratio 0.324, standard error (SE) 0.4698, p = 0.017) and increased miR-195 (β-coefficient 1.442, hazard ratio 4.229, SE 0.446, p = 0.001) predicted time to disease recurrence. Survival regression trees analysis illustrated a relative cut-off of ≤0.488 for miR-195 and a relative cut-off of >−0.218 for miR-135b; both were associated with improved disease recurrence (p < 0.001, p = 0.015). Using multivariable analysis with all targets as predictors, miR-195 (β-coefficient 3.187, SE 1.419, p = 0.025) was the sole significant independent predictor of recurrence. Conclusion: MiR-195 has strong value in predicting time to recurrence in CRC patients. Additionally, miR-21 and miR-135b predict the degree nodal burden. Future studies may include these findings to personalize therapeutic and surgical decision making.
Collapse
Affiliation(s)
- Matthew G. Davey
- Department of Surgery, Lambe Institute for Translational Research, National University of Ireland, H91 YR71 Galway, Ireland; (G.F.); (H.A.); (A.J.L.); (M.J.K.); (N.M.)
- Correspondence:
| | - Gerard Feeney
- Department of Surgery, Lambe Institute for Translational Research, National University of Ireland, H91 YR71 Galway, Ireland; (G.F.); (H.A.); (A.J.L.); (M.J.K.); (N.M.)
| | - Heidi Annuk
- Department of Surgery, Lambe Institute for Translational Research, National University of Ireland, H91 YR71 Galway, Ireland; (G.F.); (H.A.); (A.J.L.); (M.J.K.); (N.M.)
| | - Maxwell Paganga
- School of Mathematical and Statistical Sciences, National University of Ireland, H91 H3CY Galway, Ireland; (M.P.); (E.H.)
| | - Emma Holian
- School of Mathematical and Statistical Sciences, National University of Ireland, H91 H3CY Galway, Ireland; (M.P.); (E.H.)
| | - Aoife J. Lowery
- Department of Surgery, Lambe Institute for Translational Research, National University of Ireland, H91 YR71 Galway, Ireland; (G.F.); (H.A.); (A.J.L.); (M.J.K.); (N.M.)
| | - Michael J. Kerin
- Department of Surgery, Lambe Institute for Translational Research, National University of Ireland, H91 YR71 Galway, Ireland; (G.F.); (H.A.); (A.J.L.); (M.J.K.); (N.M.)
| | - Nicola Miller
- Department of Surgery, Lambe Institute for Translational Research, National University of Ireland, H91 YR71 Galway, Ireland; (G.F.); (H.A.); (A.J.L.); (M.J.K.); (N.M.)
| |
Collapse
|
29
|
Cellular senescence in the Aging Brain: A promising target for neurodegenerative diseases. Mech Ageing Dev 2022; 204:111675. [DOI: 10.1016/j.mad.2022.111675] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/24/2022] [Accepted: 04/07/2022] [Indexed: 01/10/2023]
|
30
|
Cai Y, Yang Y, Zhang X, Ma Q, Li M. TRPM2-AS promotes the malignancy of osteosarcoma cells by targeting miR-15b-5p/PPM1D axis. Cell Cycle 2022; 21:835-850. [PMID: 35100080 PMCID: PMC8973373 DOI: 10.1080/15384101.2022.2033414] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Osteosarcoma (OS) is a malignant tumor with a low survival rate and a high incidence rate worldwide. Although research has reported the involvement of long non-coding RNAs (lncRNAs) in the pathogenesis of OS cells, the role of TRPM2-AS, miR-15b-5p, and PPM1D in OS progression remains unclear. This study aimed to examine the interaction of the TRPM2-AS/miR-15b-5p/PPM1D axis in OS cells to gain new insights into the molecular mechanism and pathogenesis of OS. After performing in vitro functional assays, we discovered that TRPM2-AS was overexpressed in OS cells. TRPM2-AS silencing impaired OS cell viability, proliferation, and migration, while it induced apoptosis in OS cells in vitro. Our experimental analysis also revealed that PPM1D is a direct target of miR-15b-5p. TRPM2-AS silencing was found to reverse the tumorigenic effect of the miR-15b-5p inhibitor, while the miR-15b-5p inhibitor restored the inhibition of OS caused by silencing PPM1D. Moreover, our findings revealed that miR-15b-5p exerted its tumor-suppressive role by directly targeting PPM1D. In conclusion, this study suggests that TRPM2-AS could promote OS cell malignancy by sponging miR-15b-5p/PPM1D axis.
Collapse
Affiliation(s)
- Yingchun Cai
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,CONTACT Yingchun Cai Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou450052, Henan, China
| | - Yudan Yang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xudong Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingqing Ma
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengyi Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
31
|
Jiang L, Ingelshed K, Shen Y, Boddul SV, Iyer VS, Kasza Z, Sedimbi S, Lane DP, Wermeling F. CRISPR/Cas9-Induced DNA Damage Enriches for Mutations in a p53-Linked Interactome: Implications for CRISPR-Based Therapies. Cancer Res 2022; 82:36-45. [PMID: 34750099 PMCID: PMC9397613 DOI: 10.1158/0008-5472.can-21-1692] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/08/2021] [Accepted: 10/20/2021] [Indexed: 01/07/2023]
Abstract
Inactivating p53 mutations are the most abundant genetic alterations found in cancer. Here we show that CRISPR/Cas9-induced double-stranded DNA breaks enrich for cells deficient in p53 and in genes of a core CRISPR-p53 tumor suppressor interactome. Such enrichment could predispose to cancer development and thus pose a challenge for clinical CRISPR use. Transient p53 inhibition could suppress the enrichment of cells with these mutations. The level of DNA damage response induced by an sgRNA influenced the enrichment of p53-deficient cells and could be a relevant parameter in sgRNA design to limit cellular enrichment. Furthermore, a dataset of >800 human cancer cell lines identified additional factors influencing the enrichment of p53-mutated cells, including strong baseline CDKN1A expression as a predictor for an active CRISPR-p53 axis. Taken together, these data provide details about p53 biology in the context of CRISPR-induced DNA damage and identify strategies to enable safer CRISPR use. SIGNIFICANCE: CRISPR-mediated DNA damage enriches for cells with escape mutations in a core CRISPR-p53 interactome, which can be suppressed by transient inhibition of p53.
Collapse
Affiliation(s)
- Long Jiang
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Katrine Ingelshed
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Yunbing Shen
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Sanjaykumar V. Boddul
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Vaishnavi Srinivasan Iyer
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden.,School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore
| | - Zsolt Kasza
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Saikiran Sedimbi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - David P. Lane
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,p53 Laboratory (p53Lab), Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Fredrik Wermeling
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden.,Corresponding Author: Fredrik Wermeling, Department of Medicine, Solna, Karolinska Institutet, Stockholm 17176, Sweden. E-mail:
| |
Collapse
|
32
|
Abstract
Gadd45a, Gadd45b, and Gadd45g have been implicated in cell cycle arrest, DNA repair, apoptosis, innate immunity, genomic stability, and more recently in senescence. Evidence has accumulated that Gadd45a deficiency results in escape of mouse embryo fibroblasts from senescence, whereas Gadd45b deficiency promotes premature senescence and skin aging. Moreover, recently Gadd45b deficiency was found to promote senescence and attenuate liver fibrosis, whereas Gadd45a was observed to exert a protective effect against hepatic fibrosis. These findings indicate that the Gadd45 stress response proteins play important roles in modulating cellular responses to senescence. Thus, exploring how Gadd45 proteins modulate cellular senescence has the potential to provide new and innovative tools to treat cancer as well as liver disease.
Collapse
Affiliation(s)
- M Raza Zaidi
- Fels Cancer Institute for Personalized Medicine, Department of Cancer and Cellular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.
| | - Dan A Liebermann
- Fels Cancer Institute for Personalized Medicine, Department of Cancer and Cellular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
33
|
The p38 MAPK Components and Modulators as Biomarkers and Molecular Targets in Cancer. Int J Mol Sci 2021; 23:ijms23010370. [PMID: 35008796 PMCID: PMC8745478 DOI: 10.3390/ijms23010370] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/27/2021] [Accepted: 12/27/2021] [Indexed: 02/07/2023] Open
Abstract
The mitogen-activated protein kinase (MAPK) family is an important bridge in the transduction of extracellular and intracellular signals in different responses at the cellular level. Within this MAPK family, the p38 kinases can be found altered in various diseases, including cancer, where these kinases play a fundamental role, sometimes with antagonistic mechanisms of action, depending on several factors. In fact, this family has an immense number of functionalities, many of them yet to be discovered in terms of regulation and action in different types of cancer, being directly involved in the response to cancer therapies. To date, three main groups of MAPKs have been identified in mammals: the extracellular signal-regulated kinases (ERK), Jun N-terminal kinase (JNK), and the different isoforms of p38 (α, β, γ, δ). In this review, we highlight the mechanism of action of these kinases, taking into account their extensive regulation at the cellular level through various modifications and modulations, including a wide variety of microRNAs. We also analyze the importance of the different isoforms expressed in the different tissues and their possible role as biomarkers and molecular targets. In addition, we include the latest preclinical and clinical trials with different p38-related drugs that are ongoing with hopeful expectations in the present/future of developing precision medicine in cancer.
Collapse
|
34
|
Traub B, Roth A, Kornmann M, Knippschild U, Bischof J. Stress-activated kinases as therapeutic targets in pancreatic cancer. World J Gastroenterol 2021; 27:4963-4984. [PMID: 34497429 PMCID: PMC8384741 DOI: 10.3748/wjg.v27.i30.4963] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/17/2021] [Accepted: 07/20/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is a dismal disease with high incidence and poor survival rates. With the aim to improve overall survival of pancreatic cancer patients, new therapeutic approaches are urgently needed. Protein kinases are key regulatory players in basically all stages of development, maintaining physiologic functions but also being involved in pathogenic processes. c-Jun N-terminal kinases (JNK) and p38 kinases, representatives of the mitogen-activated protein kinases, as well as the casein kinase 1 (CK1) family of protein kinases are important mediators of adequate response to cellular stress following inflammatory and metabolic stressors, DNA damage, and others. In their physiologic roles, they are responsible for the regulation of cell cycle progression, cell proliferation and differentiation, and apoptosis. Dysregulation of the underlying pathways consequently has been identified in various cancer types, including pancreatic cancer. Pharmacological targeting of those pathways has been the field of interest for several years. While success in earlier studies was limited due to lacking specificity and off-target effects, more recent improvements in small molecule inhibitor design against stress-activated protein kinases and their use in combination therapies have shown promising in vitro results. Consequently, targeting of JNK, p38, and CK1 protein kinase family members may actually be of particular interest in the field of precision medicine in patients with highly deregulated kinase pathways related to these kinases. However, further studies are warranted, especially involving in vivo investigation and clinical trials, in order to advance inhibition of stress-activated kinases to the field of translational medicine.
Collapse
Affiliation(s)
- Benno Traub
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm 89081, Germany
| | - Aileen Roth
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm 89081, Germany
| | - Marko Kornmann
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm 89081, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm 89081, Germany
| | - Joachim Bischof
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm 89081, Germany
| |
Collapse
|
35
|
Yang Z, Sun H, Ma W, Wu K, Peng G, Ou T, Wu S. Down-regulation of Polo-like kinase 4 (PLK4) induces G1 arrest via activation of the p38/p53/p21 signalling pathway in bladder cancer. FEBS Open Bio 2021; 11:2631-2646. [PMID: 34342940 PMCID: PMC8409300 DOI: 10.1002/2211-5463.13262] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 06/22/2021] [Accepted: 08/02/2021] [Indexed: 12/24/2022] Open
Abstract
Polo-like kinase 4 (PLK4) has been reported to contribute to tumor growth, invasion, and metastasis. However, the role of PLK4 in human bladder cancer (BC) remains unclear. Here, we demonstrate the regulatory function of PLK4 in human BC progression. PLK4 is overexpressed in BC cell lines and tissues, and its overexpression correlated with poor prognosis. Our transcriptome analysis combined with subsequent functional assays indicated that PLK4 inhibition can suppress BC cell growth and induce cell cycle arrest at G1 phase via activation of the p38/p53/p21 pathway in vitro and in vivo. Overall, our data suggest that PLK4 is a critical regulator of BC cell proliferation, and thus it may have potential as a novel molecular target for BC treatment.
Collapse
Affiliation(s)
- Ziyi Yang
- Shenzhen University Health Science Center, Shenzhen, Guangdong province, China.,Institute of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518000, Guangdong province, China
| | - Haiyan Sun
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518000, Guangdong province, China
| | - Wenlong Ma
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518000, Guangdong province, China
| | - Kai Wu
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518000, Guangdong province, China
| | - Guoyu Peng
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518000, Guangdong province, China
| | - Tong Ou
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518000, Guangdong province, China
| | - Song Wu
- Shenzhen University Health Science Center, Shenzhen, Guangdong province, China.,Institute of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518000, Guangdong province, China
| |
Collapse
|
36
|
Storchova R, Burdova K, Palek M, Medema RH, Macurek L. A novel assay for screening WIP1 phosphatase substrates in nuclear extracts. FEBS J 2021; 288:6035-6051. [PMID: 33982878 DOI: 10.1111/febs.15965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/13/2021] [Accepted: 05/10/2021] [Indexed: 11/30/2022]
Abstract
Upon exposure to genotoxic stress, cells activate DNA damage response (DDR) that coordinates DNA repair with a temporal arrest in the cell cycle progression. DDR is triggered by activation of ataxia telangiectasia mutated/ataxia telangiectasia and Rad3-related protein kinases that phosphorylate multiple targets including tumor suppressor protein tumor suppressor p53 (p53). In addition, DNA damage can activate parallel stress response pathways [such as mitogen-activated protein kinase p38 alpha (p38)/MAPK-activated protein kinase 2 (MK2) kinases] contributing to establishing the cell cycle arrest. Wild-type p53-induced phosphatase 1 (WIP1) controls timely inactivation of DDR and is needed for recovery from the G2 checkpoint by counteracting the function of p53. Here, we developed a simple in vitro assay for testing WIP1 substrates in nuclear extracts. Whereas we did not detect any activity of WIP1 toward p38/MK2, we confirmed p53 as a substrate of WIP1. Inhibition or inactivation of WIP1 in U2OS cells increased phosphorylation of p53 at S15 and potentiated its acetylation at K382. Further, we identified Deleted in breast cancer gene 1 (DBC1) as a new substrate of WIP1 but surprisingly, depletion of DBC1 did not interfere with the ability of WIP1 to regulate p53 acetylation. Instead, we have found that WIP1 activity suppresses p53-K382 acetylation by inhibiting the interaction between p53 and the acetyltransferase p300. Newly established phosphatase assay allows an easy comparison of WIP1 ability to dephosphorylate various proteins and thus contributes to identification of its physiological substrates.
Collapse
Affiliation(s)
- Radka Storchova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.,Faculty of Science, Charles University, Prague, Czech Republic
| | - Kamila Burdova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Matous Palek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - René H Medema
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Libor Macurek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
37
|
Eren MK, Kartal NB, Pilevneli H. Oncogenic WIP1 phosphatase attenuates the DNA damage response and sensitizes p53 mutant Jurkat cells to apoptosis. Oncol Lett 2021; 21:479. [PMID: 33968195 PMCID: PMC8100942 DOI: 10.3892/ol.2021.12740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/25/2021] [Indexed: 12/19/2022] Open
Abstract
Wild-type (wt) p53-induced phosphatase 1 (Wip1), encoded by the protein phosphatase, Mg2+/Mn2+ dependent 1D (PPM1D) gene, is a serine/threonine phosphatase induced upon genotoxic stress in a p53-dependent manner. Wip1/PPM1D is frequently overexpressed, amplified and mutated in human solid tumors harboring wt p53 and is thus currently recognized as an oncogene. Oncogenic Wip1 dampens cellular stress responses, such as cell cycle checkpoints, apoptosis and senescence, and consequently increases resistance to anticancer therapeutics. Targeting Wip1 has emerged as a therapeutic strategy for tumors harboring wt p53. However, little is known about the efficacy of Wip1-targeted therapies in tumors lacking p53. The present study aimed to investigate the potential role of oncogenic Wip1 in p53 mutant (mt) Jurkat cells. In the present study, it was demonstrated that p53 mt Jurkat cells exhibited PPM1D/Wip1 gene amplification and expressed relatively high levels of Wip1, as confirmed by gene copy number and RNA expression analysis. In addition, Jurkat cells underwent G2 cell cycle arrest, apoptotic cell death and senescence in response to etoposide and doxorubicin, although the phosphorylation levels of DNA damage response (DDR) elements, including ataxia-telangiectasia mutated, ataxia-telangiestasia and Rad3-related, checkpoint kinase (Chk)1 and Chk2 were significantly low. Accordingly, the targeting of Wip1 phosphatase by RNA interference increased the phosphorylation of DDR elements, but decreased the rate of apoptosis in response to etoposide or doxorubicin in Jurkat cells. The induction of senescence or cell cycle arrest was not affected by the knockdown of Wip1. The results suggest that increased Wip1 expression enhances the apoptotic sensitivity of Jurkat cells in response to chemotherapeutic agents by attenuating DDR signaling. The present study highlights the possible pro-apoptotic role of Wip1 in a p53 mt T-cell acute lymphoblastic leukemia cell line. The data suggest the careful consideration of future treatment strategies aiming to manipulate or target Wip1 in human cancers lacking p53.
Collapse
Affiliation(s)
- Mehtap Kilic Eren
- Department of Medical Biology, Faculty of Medicine, University of Aydin Adnan Menderes, 09010 Aydin, Turkey
| | - Nur Betül Kartal
- Department of Medical Biochemistry, Institute of Health Sciences, University of Aydin Adnan Menderes, 09010 Aydin, Turkey
| | - Hatice Pilevneli
- Department of Medical Biology, Institute of Health Sciences, University of Aydin Adnan Menderes, 09010 Aydin, Turkey
| |
Collapse
|
38
|
Zhang X, Park JE, Kim EH, Hong J, Hwang KT, Kim YA, Jang CY. Wip1 controls the translocation of the chromosomal passenger complex to the central spindle for faithful mitotic exit. Cell Mol Life Sci 2021; 78:2821-2838. [PMID: 33067654 PMCID: PMC11072438 DOI: 10.1007/s00018-020-03665-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 08/12/2020] [Accepted: 10/05/2020] [Indexed: 10/23/2022]
Abstract
Dramatic cellular reorganization in mitosis critically depends on the timely and temporal phosphorylation of a broad range of proteins, which is mediated by the activation of the mitotic kinases and repression of counteracting phosphatases. The mitosis-to-interphase transition, which is termed mitotic exit, involves the removal of mitotic phosphorylation by protein phosphatases. Although protein phosphatase 1 (PP1) and protein phosphatase 2A (PP2A) drive this reversal in animal cells, the phosphatase network associated with ordered bulk dephosphorylation in mitotic exit is not fully understood. Here, we describe a new mitotic phosphatase relay in which Wip1/PPM1D phosphatase activity is essential for chromosomal passenger complex (CPC) translocation to the anaphase central spindle after release from the chromosome via PP1-mediated dephosphorylation of histone H3T3. Depletion of endogenous Wip1 and overexpression of the phosphatase-dead mutant disturbed CPC translocation to the central spindle, leading to failure of cytokinesis. While Wip1 was degraded in early mitosis, its levels recovered in anaphase and the protein functioned as a Cdk1-counteracting phosphatase at the anaphase central spindle and midbody. Mechanistically, Wip1 dephosphorylated Thr-59 in inner centromere protein (INCENP), which, subsequently bound to MKLP2 and recruited other components to the central spindle. Furthermore, Wip1 overexpression is associated with the overall survival rate of patients with breast cancer, suggesting that Wip1 not only functions as a weak oncogene in the DNA damage network but also as a tumor suppressor in mitotic exit. Altogether, our findings reveal that sequential dephosphorylation of mitotic phosphatases provides spatiotemporal regulation of mitotic exit to prevent tumor initiation and progression.
Collapse
Affiliation(s)
- Xianghua Zhang
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Ji Eun Park
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Eun Ho Kim
- Department of Biochemistry, School of Medicine, Catholic University of Daegu, Daegu, 42472, Republic of Korea
| | - Jihee Hong
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Ki-Tae Hwang
- Department of Surgery, Seoul National University Boramae Medical Center, Seoul, 07061, Republic of Korea
| | - Young A Kim
- Department of Pathology, Seoul National University Boramae Medical Center, Seoul, 07061, Republic of Korea.
| | - Chang-Young Jang
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| |
Collapse
|
39
|
Systematic characterization of mutations altering protein degradation in human cancers. Mol Cell 2021; 81:1292-1308.e11. [PMID: 33567269 PMCID: PMC9245451 DOI: 10.1016/j.molcel.2021.01.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 12/01/2020] [Accepted: 01/17/2021] [Indexed: 02/06/2023]
Abstract
The ubiquitin-proteasome system (UPS) is the primary route for selective protein degradation in human cells. The UPS is an attractive target for novel cancer therapies, but the precise UPS genes and substrates important for cancer growth are incompletely understood. Leveraging multi-omics data across more than 9,000 human tumors and 33 cancer types, we found that over 19% of all cancer driver genes affect UPS function. We implicate transcription factors as important substrates and show that c-Myc stability is modulated by CUL3. Moreover, we developed a deep learning model (deepDegron) to identify mutations that result in degron loss and experimentally validated the prediction that gain-of-function truncating mutations in GATA3 and PPM1D result in increased protein stability. Last, we identified UPS driver genes associated with prognosis and the tumor microenvironment. This study demonstrates the important role of UPS dysregulation in human cancer and underscores the potential therapeutic utility of targeting the UPS.
Collapse
|
40
|
Phosphatase magnesium-dependent 1 δ (PPM1D), serine/threonine protein phosphatase and novel pharmacological target in cancer. Biochem Pharmacol 2020; 184:114362. [PMID: 33309518 DOI: 10.1016/j.bcp.2020.114362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/20/2022]
Abstract
Aberrations in DNA damage response genes are recognized mediators of tumorigenesis and resistance to chemo- and radiotherapy. While protein phosphatase magnesium-dependent 1 δ (PPM1D), located on the long arm of chromosome 17 at 17q22-23, is a key regulator of cellular responses to DNA damage, amplification, overexpression, or mutation of this gene is important in a wide range of pathologic processes. In this review, we describe the physiologic function of PPM1D, as well as its role in diverse processes, including fertility, development, stemness, immunity, tumorigenesis, and treatment responsiveness. We highlight both the advances and limitations of current approaches to targeting malignant processes mediated by pathogenic alterations in PPM1D with the goal of providing rationale for continued research and development of clinically viable treatment approaches for PPM1D-associated diseases.
Collapse
|
41
|
Husby S, Hjermind Justesen E, Grønbæk K. Protein phosphatase, Mg 2+/Mn 2+-dependent 1D (PPM1D) mutations in haematological cancer. Br J Haematol 2020; 192:697-705. [PMID: 33616916 DOI: 10.1111/bjh.17120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/02/2020] [Indexed: 01/07/2023]
Abstract
Until recently, the protein phosphatase, Mg2+/Mn2+-dependent 1D (PPM1D) gene had not been examined in haematological cancer, but several studies have now explored the functional role of this gene and its aberrations. It is often mutated in the context of clonal haemopoiesis (including in patients with lymphoma, myeloproliferative neoplasms and myelodysplastic syndrome) and mutations have been associated with exposure to cytotoxic and radiation therapy, development of therapy-related neoplasms and inferior survival. The vast majority of PPM1D mutations found in haematopoietic cells are of the nonsense or frameshift type and located within terminal exon 6. These genetic defects are rarely found in the blood of healthy individuals. PPM1D encodes the PPM1D phosphatase [also named wild-type p53-induced phosphatase 1 (WIP1)], which negatively regulates signalling molecules within the DNA damage response pathway, including tumour suppressor p53. Clonal expansion of PPM1D mutant haematopoietic cells can potentially be prevented with inhibitors; however, human trials are awaited. In the present review, we provide a review of the literature regarding PPM1D and its role in haematological cancer.
Collapse
Affiliation(s)
- Simon Husby
- Department of Haematology, Rigshospitalet, Copenhagen, Denmark.,Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Emma Hjermind Justesen
- Department of Haematology, Rigshospitalet, Copenhagen, Denmark.,Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Kirsten Grønbæk
- Department of Haematology, Rigshospitalet, Copenhagen, Denmark.,Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
42
|
Ren XH, He XY, Liu BY, Xu C, Cheng SX. Self-Assembled Plasmid Delivery System for PPM1D Knockout to Reverse Tumor Malignancy. ACS APPLIED BIO MATERIALS 2020; 3:7831-7839. [DOI: 10.1021/acsabm.0c01009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Xiao-He Ren
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, People’s Republic of China
| | - Xiao-Yan He
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, People’s Republic of China
| | - Bo-Ya Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, People’s Republic of China
| | - Chang Xu
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, People’s Republic of China
| | - Si-Xue Cheng
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, People’s Republic of China
| |
Collapse
|
43
|
Nisar S, Hashem S, Macha MA, Yadav SK, Muralitharan S, Therachiyil L, Sageena G, Al-Naemi H, Haris M, Bhat AA. Exploring Dysregulated Signaling Pathways in Cancer. Curr Pharm Des 2020; 26:429-445. [PMID: 31939726 DOI: 10.2174/1381612826666200115095937] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 11/27/2019] [Indexed: 02/08/2023]
Abstract
Cancer cell biology takes advantage of identifying diverse cellular signaling pathways that are disrupted in cancer. Signaling pathways are an important means of communication from the exterior of cell to intracellular mediators, as well as intracellular interactions that govern diverse cellular processes. Oncogenic mutations or abnormal expression of signaling components disrupt the regulatory networks that govern cell function, thus enabling tumor cells to undergo dysregulated mitogenesis, to resist apoptosis, and to promote invasion to neighboring tissues. Unraveling of dysregulated signaling pathways may advance the understanding of tumor pathophysiology and lead to the improvement of targeted tumor therapy. In this review article, different signaling pathways and how their dysregulation contributes to the development of tumors have been discussed.
Collapse
Affiliation(s)
- Sabah Nisar
- Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Sheema Hashem
- Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Muzafar A Macha
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States.,Department of Biotechnology, Central University of Kashmir, Ganderbal, Jammu and Kashmir, India
| | - Santosh K Yadav
- Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | | | - Lubna Therachiyil
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | | | - Hamda Al-Naemi
- Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Mohammad Haris
- Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar.,Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Ajaz A Bhat
- Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| |
Collapse
|
44
|
Martinikova AS, Burocziova M, Stoyanov M, Macurek L. Truncated PPM1D Prevents Apoptosis in the Murine Thymus and Promotes Ionizing Radiation-Induced Lymphoma. Cells 2020; 9:cells9092068. [PMID: 32927737 PMCID: PMC7565556 DOI: 10.3390/cells9092068] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/04/2020] [Accepted: 09/06/2020] [Indexed: 12/20/2022] Open
Abstract
Genome integrity is protected by the cell-cycle checkpoints that prevent cell proliferation in the presence of DNA damage and allow time for DNA repair. The transient checkpoint arrest together with cellular senescence represent an intrinsic barrier to tumorigenesis. Tumor suppressor p53 is an integral part of the checkpoints and its inactivating mutations promote cancer growth. Protein phosphatase magnesium-dependent 1 (PPM1D) is a negative regulator of p53. Although its loss impairs recovery from the G2 checkpoint and promotes induction of senescence, amplification of the PPM1D locus or gain-of-function truncating mutations of PPM1D occur in various cancers. Here we used a transgenic mouse model carrying a truncating mutation in exon 6 of PPM1D (Ppm1dT). As with human cell lines, we found that the truncated PPM1D was present at high levels in the mouse thymus. Truncated PPM1D did not affect differentiation of T-cells in the thymus but it impaired their response to ionizing radiation (IR). Thymocytes in Ppm1dT/+ mice did not arrest in the checkpoint and continued to proliferate despite the presence of DNA damage. In addition, we observed a decreased level of apoptosis in the thymi of Ppm1dT/+ mice. Moreover, the frequency of the IR-induced T-cell lymphomas increased in Ppm1dT/+Trp53+/- mice resulting in decreased survival. We conclude that truncated PPM1D partially suppresses the p53 pathway in the mouse thymus and potentiates tumor formation under the condition of a partial loss of p53 function.
Collapse
Affiliation(s)
- Andra S. Martinikova
- Laboratory of Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, CZ14220 Prague, Czech Republic; (A.S.M.); (M.B.); (M.S.)
- Department of Developmental and Cell Biology, Faculty of Science, Charles University, Albertov 6, CZ12800 Prague, Czech Republic
| | - Monika Burocziova
- Laboratory of Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, CZ14220 Prague, Czech Republic; (A.S.M.); (M.B.); (M.S.)
| | - Miroslav Stoyanov
- Laboratory of Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, CZ14220 Prague, Czech Republic; (A.S.M.); (M.B.); (M.S.)
| | - Libor Macurek
- Laboratory of Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, CZ14220 Prague, Czech Republic; (A.S.M.); (M.B.); (M.S.)
- Correspondence: ; Tel.: +42-(0)2-4106-3210
| |
Collapse
|
45
|
TRIM37 controls cancer-specific vulnerability to PLK4 inhibition. Nature 2020; 585:440-446. [PMID: 32908304 PMCID: PMC7501188 DOI: 10.1038/s41586-020-2710-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 06/09/2020] [Indexed: 12/21/2022]
Abstract
Centrosomes catalyze microtubule formation for mitotic spindle assembly1. Centrosomes duplicate once per cell cycle in a process controlled the kinase PLK42,3. Following chemical PLK4 inhibition, cell division in the absence of centrosome duplication generates centrosome-less cells that exhibit delayed, acentrosomal spindle assembly4. Whether PLK4 inhibitors can be leveraged for cancer treatment is not yet clear. Here, we show that acentrosomal spindle assembly following PLK4 inhibition depends on levels of the centrosomal ubiquitin ligase TRIM37. Low TRIM37 accelerates acentrosomal spindle assembly and improves proliferation following PLK4 inhibition, whereas high TRIM37 inhibits acentrosomal spindle assembly, leading to mitotic failure and cessation of proliferation. The Chr17q region containing the TRIM37 gene is frequently amplified in neuroblastoma and in breast cancer5–8, which renders these cancer types highly sensitive to PLK4 inhibition. TRIM37 inactivation improves acentrosomal mitosis because TRIM37 prevents PLK4 self-assembly into centrosome-independent condensates that serve as ectopic microtubule-organizing centers. By contrast, elevated TRIM37 expression inhibits acentrosomal spindle assembly via a distinct mechanism that involves degradation of the centrosomal component CEP192. Thus, TRIM37 is a critical determinant of mitotic vulnerability to PLK4 inhibition. Linkage of TRIM37 to prevalent cancer-associated genomic changes, including 17q gain in neuroblastoma and 17q23 amplification in breast cancer, may offer an opportunity to use PLK4 inhibition to trigger selective mitotic failure and provide new avenues to treatments for these cancers.
Collapse
|
46
|
Hat B, Jaruszewicz-Błońska J, Lipniacki T. Model-based optimization of combination protocols for irradiation-insensitive cancers. Sci Rep 2020; 10:12652. [PMID: 32724100 PMCID: PMC7387345 DOI: 10.1038/s41598-020-69380-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 06/19/2020] [Indexed: 01/07/2023] Open
Abstract
Alternations in the p53 regulatory network may render cancer cells resistant to the radiation-induced apoptosis. In this theoretical study we search for the best protocols combining targeted therapy with radiation to treat cancers with wild-type p53, but having downregulated expression of PTEN or overexpression of Wip1 resulting in resistance to radiation monotherapy. Instead of using the maximum tolerated dose paradigm, we exploit stochastic computational model of the p53 regulatory network to calculate apoptotic fractions for both normal and cancer cells. We consider combination protocols, with irradiations repeated every 12, 18, 24, or 36 h to find that timing between Mdm2 inhibitor delivery and irradiation significantly influences the apoptotic cell fractions. We assume that uptake of the inhibitor is higher by cancer than by normal cells and that cancer cells receive higher irradiation doses from intersecting beams. These two assumptions were found necessary for the existence of protocols inducing massive apoptosis in cancer cells without killing large fraction of normal cells neighboring tumor. The best found protocols have irradiations repeated every 24 or 36 h with two inhibitor doses per irradiation cycle, and allow to induce apoptosis in more than 95% of cancer cells, killing less than 10% of normal cells.
Collapse
Affiliation(s)
- Beata Hat
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
| | | | - Tomasz Lipniacki
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
47
|
Shi L, Tian Q, Feng C, Zhang P, Zhao Y. The biological function and the regulatory roles of wild-type p53-induced phosphatase 1 in immune system. Int Rev Immunol 2020; 39:280-291. [PMID: 32696682 DOI: 10.1080/08830185.2020.1795153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Wild-type p53-induced phosphatase 1 (WIP1) belongs to the protein phosphatase 2C (PP2C) family and is a mammalian serine/threonine specific protein phosphatase to dephosphorylate numerous signaling molecules. Mammalian WIP1 regulates a wide array of targeting molecules and plays key regulatory roles in many cell processes such as DNA damage and repair, cell proliferation, differentiation, apoptosis, and senescence. WIP1 promotes the formation and development of tumors as an oncogene and a negative regulator of p53. It is also involved in the regulation of aging, neurological diseases and immune diseases. Recent studies demonstrated the critical roles of WIP1 in the differentiation and function of immune cells including T cells, neutrophils and macrophages. In the present manuscript, we briefly summarized the expression patterns, biological function and the target molecules and signal pathways of WIP1 and mainly discussed the latest advances on the regulatory effects of WIP1 in the immune system. WIP1 may be a potential target molecule to treat cancers and immune diseases such as allergic asthma.
Collapse
Affiliation(s)
- Lu Shi
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qianchuan Tian
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chang Feng
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Peng Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
48
|
Ozen M, Lipniacki T, Levchenko A, Emamian ES, Abdi A. Modeling and measurement of signaling outcomes affecting decision making in noisy intracellular networks using machine learning methods. Integr Biol (Camb) 2020; 12:122-138. [PMID: 32424393 DOI: 10.1093/intbio/zyaa009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 12/30/2022]
Abstract
Characterization of decision-making in cells in response to received signals is of importance for understanding how cell fate is determined. The problem becomes multi-faceted and complex when we consider cellular heterogeneity and dynamics of biochemical processes. In this paper, we present a unified set of decision-theoretic, machine learning and statistical signal processing methods and metrics to model the precision of signaling decisions, in the presence of uncertainty, using single cell data. First, we introduce erroneous decisions that may result from signaling processes and identify false alarms and miss events associated with such decisions. Then, we present an optimal decision strategy which minimizes the total decision error probability. Additionally, we demonstrate how graphing receiver operating characteristic curves conveniently reveals the trade-off between false alarm and miss probabilities associated with different cell responses. Furthermore, we extend the introduced framework to incorporate the dynamics of biochemical processes and reactions in a cell, using multi-time point measurements and multi-dimensional outcome analysis and decision-making algorithms. The introduced multivariate signaling outcome modeling framework can be used to analyze several molecular species measured at the same or different time instants. We also show how the developed binary outcome analysis and decision-making approach can be extended to more than two possible outcomes. As an example and to show how the introduced methods can be used in practice, we apply them to single cell data of PTEN, an important intracellular regulatory molecule in a p53 system, in wild-type and abnormal cells. The unified signaling outcome modeling framework presented here can be applied to various organisms ranging from viruses, bacteria, yeast and lower metazoans to more complex organisms such as mammalian cells. Ultimately, this signaling outcome modeling approach can be utilized to better understand the transition from physiological to pathological conditions such as inflammation, various cancers and autoimmune diseases.
Collapse
Affiliation(s)
- Mustafa Ozen
- Center for Wireless Information Processing, Department of Electrical and Computer Engineering, New Jersey Institute of Technology, 323 King Blvd, Newark, NJ 07102, USA
| | - Tomasz Lipniacki
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawinskiego 5B, 02-106 Warsaw, Poland
| | - Andre Levchenko
- Yale Systems Biology Institute and Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Effat S Emamian
- Advanced Technologies for Novel Therapeutics, Enterprise Development Center, New Jersey Institute of Technology, 211 Warren St., Newark, NJ 07103, USA
| | - Ali Abdi
- Center for Wireless Information Processing, Department of Electrical and Computer Engineering, New Jersey Institute of Technology, 323 King Blvd, Newark, NJ 07102, USA.,Department of Biological Sciences, New Jersey Institute of Technology, 323 King Blvd, Newark, NJ 07102, USA
| |
Collapse
|
49
|
Deng W, Li J, Dorrah K, Jimenez-Tapia D, Arriaga B, Hao Q, Cao W, Gao Z, Vadgama J, Wu Y. The role of PPM1D in cancer and advances in studies of its inhibitors. Biomed Pharmacother 2020; 125:109956. [PMID: 32006900 PMCID: PMC7080581 DOI: 10.1016/j.biopha.2020.109956] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/08/2020] [Accepted: 01/23/2020] [Indexed: 12/16/2022] Open
Abstract
A greater understanding of factors causing cancer initiation, progression and evolution is of paramount importance. Among them, the serine/threonine phosphatase PPM1D, also referred to as wild-type p53-induced phosphatase 1 (Wip1) or protein phosphatase 2C delta (PP2Cδ), is emerging as an important oncoprotein due to its negative regulation on a number of crucial cancer suppressor pathways. Initially identified as a p53-regulated gene, PPM1D has been afterwards found amplified and more recently mutated in many human cancers such as breast cancer. The latest progress in this field further reveals that selective inhibition of PPM1D to delay tumor onset or reduce tumor burden represents a promising anti-cancer strategy. Here, we review the advances in the studies of the PPM1D activity and its relevance to various cancers, and recent progress in development of PPM1D inhibitors and discuss their potential application in cancer therapy. Consecutive research on PPM1D and its relationship with cancer is essential, as it ultimately contributes to the etiology and treatment of cancer.
Collapse
Affiliation(s)
- Wenhong Deng
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Jieqing Li
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Kimberly Dorrah
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Denise Jimenez-Tapia
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Brando Arriaga
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Qiongyu Hao
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Wei Cao
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Zhaoxia Gao
- Department of General Surgery, 5th Hospital of Wuhan, Wuhan, 430050, China; Department of Surgery, Johns Hopkins Hospital Bayview Campus, Baltimore, MD, USA
| | - Jay Vadgama
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.
| | - Yong Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.
| |
Collapse
|
50
|
Yu S, Cheng Y, Li B, Xue J, Yin Y, Gao J, Gong Z, Wang J, Mu Y. M1 macrophages accelerate renal glomerular endothelial cell senescence through reactive oxygen species accumulation in streptozotocin-induced diabetic mice. Int Immunopharmacol 2020; 81:106294. [PMID: 32062081 DOI: 10.1016/j.intimp.2020.106294] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 01/07/2023]
Abstract
Cellular senescence is a fundamental aging mechanism leading to tissue dysfunction. Accumulation of senescent cells is observed in the context of diabetes, which plays an important role in the pathogenesis of diabetes and its complications. Macrophages, the most prevalent leucocytes found in diabetic kidney, have been implicated in the modulation of cellular senescence; however, their role and mechanism in cellular senescence of diabetic kidney have not been determined. In this study, we found trends of cellular senescence in the glomeruli of streptozotocin-induced diabetic mice. The onset of glomerular senescence was confirmed by increased SA-β-gal staining, the upregulation of p16INK4a, p21, and p53 protein levels and the increased expression of SASP RNA. The senescent cells in the glomeruli were mainly endothelial cells. We next confirmed that M1 macrophages accumulated in the glomeruli, occurred just shortly before glomerular senescence. Therefore, we examined whether M1 macrophage accumulation is associated with glomerular endothelial cell senescence. Thus, an in vitro co-culture model was established using human renal glomerular endothelial cells (HRGECs) and M1-polarized THP-1 macrophages. Indeed, M1 macrophages induced senescence in HRGECs. Furthermore, intracellular ROS levels and p38 MAPK signalling activation were significantly increased in HRGECs and reducing ROS generation significantly abolished M1 macrophage-mediated endothelial senescence and p38 MAPK activation, suggesting that M1 macrophage-mediated endothelial senescence is largely dependent on ROS. Thus, our results demonstrate that kidney M1 macrophage accumulation is in connection with endothelial cell senescence and strategy to modulate M1 macrophages accumulation is promising to be a new target for immunotherapy for diabetic kidney disease and other age-related diseases.
Collapse
Affiliation(s)
- Songyan Yu
- School of Medicine, Nankai University, Tianjin, China; Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Yu Cheng
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Bing Li
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Jing Xue
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Yaqi Yin
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Jieqing Gao
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Zhengyuan Gong
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Jie Wang
- School of Medicine, Nankai University, Tianjin, China; Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Yiming Mu
- School of Medicine, Nankai University, Tianjin, China; Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China.
| |
Collapse
|