1
|
Neale DA, Morris JC, Verrills NM, Ammit AJ. Understanding the regulatory landscape of protein phosphatase 2A (PP2A): Pharmacological modulators and potential therapeutics. Pharmacol Ther 2025; 269:108834. [PMID: 40023321 DOI: 10.1016/j.pharmthera.2025.108834] [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: 10/21/2024] [Revised: 01/20/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Protein phosphatase 2A (PP2A) is a ubiquitously expressed serine/threonine phosphatase with a diverse and integral role in cellular signalling pathways. Consequently, its dysfunction is frequently observed in disease states such as cancer, inflammation and Alzheimer's disease. A growing understanding of both PP2A and its endogenous regulatory proteins has presented numerous targets for therapeutic intervention. This provides important context for the dynamic control and dysregulation of PP2A function in disease states. Understanding the intricate regulation of PP2A signalling in disease has resulted in the development of novel pharmacological agents aimed at restoring cellular homeostasis. Herein we review the structure and function of PP2A together with pharmacological modulators, both endogenous (proteins) and exogenous (small molecules and peptides), with relevance to targeting PP2A as a future pharmacotherapeutic strategy.
Collapse
Affiliation(s)
- David A Neale
- School of Chemistry, UNSW Sydney, NSW 2052, Australia
| | | | - Nicole M Verrills
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, NSW 2308, Australia; Precision Medicine Program, Hunter Medical Research Institute, New Lambton, NSW 2305, Australia
| | - Alaina J Ammit
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, Macquarie University, NSW, Australia; School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia.
| |
Collapse
|
2
|
Jian J, Yuan C, Hao H. Identifying key genes and functionally enriched pathways in acute myeloid leukemia by weighted gene co-expression network analysis. J Appl Genet 2025; 66:347-362. [PMID: 38977582 DOI: 10.1007/s13353-024-00881-0] [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: 02/23/2023] [Revised: 05/23/2024] [Accepted: 05/31/2024] [Indexed: 07/10/2024]
Abstract
Acute myeloid leukemia (AML) is characterized by the uncontrolled proliferation of myeloid leukemia cells in the bone marrow and other hematopoietic tissues and is highly heterogeneous. While with the progress of sequencing technology, understanding of the AML-related biomarkers is still incomplete. The purpose of this study is to identify potential biomarkers for prognosis of AML. Based on WGCNA analysis of gene mutation expression, methylation level distribution, mRNA expression, and AML-related genes in public databases were employed for investigating potential biomarkers for the prognosis of AML. This study screened a total of 6153 genes by analyzing various changes in 103 acute myeloid leukemia (AML) samples, including gene mutation expression, methylation level distribution, mRNA expression, and AML-related genes in public databases. Moreover, seven AML-related co-expression modules were mined by WGCNA analysis, and twelve biomarkers associated with the AML prognosis were identified from each top 10 genes of the seven co-expression modules. The AML samples were then classified into two subgroups, the prognosis of which is significantly different, based on the expression of these twelve genes. The differentially expressed 7 genes of two subgroups (HOXB-AS3, HOXB3, SLC9C2, CPNE8, MEG8, S1PR5, MIR196B) are mainly involved in glucose metabolism, glutathione biosynthesis, small G protein-mediated signal transduction, and the Rap1 signaling pathway. With the utilization of WGCNA mining, seven gene co-expression modules were identified from the TCGA database, and there are unreported genes that may be potential driver genes of AML and may be the direction to identify the possible molecular signatures to predict survival of AML patients and help guide experiments for potential clinical drug targets.
Collapse
Affiliation(s)
- Jimo Jian
- Qilu Hospital of Shandong University, Qingdao, 266035, Shandong, China
- Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Chenglu Yuan
- Qilu Hospital of Shandong University, Qingdao, 266035, Shandong, China
| | - Hongyuan Hao
- Qilu Hospital of Shandong University, Qingdao, 266035, Shandong, China.
| |
Collapse
|
3
|
Shao F, Wang R, Li X, Hu Y, Zhang Z, Cai J, Yang J, Feng X, Ren S, Huang Z, Xie Y. TTC36 promotes proliferation and drug resistance in hepatocellular carcinoma cells by inhibiting c-Myc degradation. Cell Death Dis 2025; 16:332. [PMID: 40274799 DOI: 10.1038/s41419-025-07663-4] [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: 09/10/2024] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025]
Abstract
High c-Myc protein accumulation contributes to the proliferation, invasion, and drug resistance in multiple cancer cells, but the underlying mechanism about c-Myc accumulation remains not to be elucidated. Here, we demonstrate that TTC36 promotes c-Myc protein accumulation in hepatocellular carcinoma cells, thereby driving the proliferation and sorafenib resistance in hepatocellular carcinoma cells. Ttc36 depletion disrupts the interaction between SET and PPP2R1A, consequently activating PP2A. Activated PP2A directly dephosphorylates p-c-MycS62 and activates GSK3β, relying on AKT, leading increased phosphorylation of p-c-MycT58, finally promotes FBXW7-mediated polyubiquitination and degradation of c-Myc. Inhibitors targeting GSK3β and PP2A effectively reverse the sorafenib resistance promoted by TTC36. These findings highlight the crucial role of TTC36 in c-Myc accumulation-caused proliferation and sorafenib resistance in HCC, providing a promising combination strategy for treating patients with c-Myc protein accumulation in advanced HCC.
Collapse
Affiliation(s)
- Fengling Shao
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Runzhi Wang
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xinyi Li
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yanxia Hu
- School of Life and Health Sciences, Hainan University, Haikou, China
| | - Zaikuan Zhang
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Jing Cai
- College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Jieru Yang
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xiaosong Feng
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Suxia Ren
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China.
| | - Zengyi Huang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China.
- Mitomedical laboratory of Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China.
| | - Yajun Xie
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
4
|
Ma S, Su S, Zhang X, Wang X, Yi H. CircRNA encoded-peptide: Potential stock in the transcriptomics market. Life Sci 2025; 372:123643. [PMID: 40246192 DOI: 10.1016/j.lfs.2025.123643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/03/2025] [Accepted: 04/13/2025] [Indexed: 04/19/2025]
Abstract
The emergence of circRNA-encoded peptides has sparked significant debate in recent years as a novel mode of action for circRNAs. A mounting body of evidence suggests that these peptides play vital roles in cancer development and immune responses. This review initially elucidates the presence of circRNA-encoded peptides and delineates their specific functions across various biological processes and pathological conditions. It goes on to furnish illustrative instances to underscore the pivotal involvement of circRNA-encoded peptides in both innate and adaptive immune responses. The study sheds new light on the biological roles of circRNAs, their potential tumor-promoting and tumor-suppressing functions of circRNA-encoded peptides in specific tumor environment, and their significance in immunological contexts. Meanwhile, the limitations of existing studies on circRNA-encoded peptides are discussed in depth. In particular, circRNA-encoded peptides are critically analyzed as biomarkers and therapeutic targets. Intriguingly, the review concludes with a more organized discussion of future research on circRNA-encoded peptides.
Collapse
Affiliation(s)
- Siyuan Ma
- Central Laboratory, Lequn Branch, The First Hospital of Jilin University, Changchun, Jilin 130031, China; Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, Changchun, Jilin 130021, China
| | - Sensen Su
- Central Laboratory, Lequn Branch, The First Hospital of Jilin University, Changchun, Jilin 130031, China; Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, Changchun, Jilin 130021, China; Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xiuna Zhang
- Central Laboratory, Lequn Branch, The First Hospital of Jilin University, Changchun, Jilin 130031, China; Department of Gastroenterology, Lequn Branch, The First Hospital of Jilin University, Changchun 130000, China
| | - Xiangxiu Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Gongli Hospital of Pudong New Area, Shanghai 200135, China
| | - Huanfa Yi
- Central Laboratory, Lequn Branch, The First Hospital of Jilin University, Changchun, Jilin 130031, China; Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, Changchun, Jilin 130021, China.
| |
Collapse
|
5
|
Skamagki M, Zhang C, Hacisuleyman E, Galleti G, Wu C, Vinagolu RK, Cha H, Ata D, Kim J, Weiskittel T, Diop M, Aung T, Del Latto M, Kim AS, Li Z, Miele M, Zhao R, Tang LH, Hendrickson RC, Romesser PB, Smith JJ, Giannakakou P, Darnell RB, Bott MJ, Li H, Kim K. Aging-dependent dysregulation of EXOSC2 is maintained in cancer as a dependency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647279. [PMID: 40236131 PMCID: PMC11996493 DOI: 10.1101/2025.04.04.647279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Reprogramming of aged donor tissue cells into induced pluripotent stem cells (A-iPSC) preserved the epigenetic memory of aged-donor tissue, defined as genomic instability and poor tissue differentiation in our previous study. The unbalanced expression of RNA exosome subunits affects the RNA degradation complex function and is associated with geriatric diseases including premature aging and cancer progression. We hypothesized that the age-dependent progressive subtle dysregulation of EXOSC2 (exosome component 2) causes the aging traits (abnormal cell cycle and poor tissue differentiation). We used embryonic stem cells as a tool to study EXOSC2 function as the aging trait epigenetic memory determined in A-iPSC because these aging traits could not be studied in senesced aged cells or immortalized cancer cells. We found that the regulatory subunit of PP2A phosphatase, PPP2R5E, is a key target of EXOSC2 and this regulation is preserved in stem cells and cancer.
Collapse
|
6
|
Turnham RE, Pitea A, Jang GM, Xu Z, Lim HC, Choi AL, Von Dollen J, Levin RS, Webber JT, McCarthy E, Hu J, Li X, Che L, Singh A, Yoon A, Chan G, Kelley RK, Swaney DL, Zhang W, Bandyopadhyay S, Theis FJ, Eckhardt M, Chen X, Shokat KM, Ideker T, Krogan NJ, Gordan JD. HBV Remodels PP2A Complexes to Rewire Kinase Signaling in Hepatocellular Carcinoma. Cancer Res 2025; 85:660-674. [PMID: 39652575 PMCID: PMC11949624 DOI: 10.1158/0008-5472.can-24-0456] [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: 02/06/2024] [Revised: 09/09/2024] [Accepted: 12/04/2024] [Indexed: 02/18/2025]
Abstract
Hepatitis B virus (HBV) infections promote liver cancer initiation by inducing inflammation and cellular stress. Despite a primarily indirect effect on oncogenesis, HBV is associated with a recurrent genomic phenotype in hepatocellular carcinoma (HCC), suggesting that it impacts the biology of established HCC. Characterization of the interaction of HBV with host proteins and the mechanistic contributions of HBV to HCC initiation and maintenance could provide insights into HCC biology and uncover therapeutic vulnerabilities. In this study, we used affinity purification mass spectrometry to comprehensively map a network of 145 physical interactions between HBV and human proteins in HCC. A subset of the host factors targeted by HBV proteins were preferentially mutated in non-HBV-associated HCC, suggesting that their interaction with HBV influences HCC biology. HBV interacted with proteins involved in mRNA splicing, mitogenic signaling, and DNA repair, with the latter set interacting with the HBV oncoprotein X (HBx). HBx remodeled the PP2A phosphatase complex by excluding striatin regulatory subunits from the PP2A holoenzyme, and the HBx effects on PP2A caused Hippo kinase activation. In parallel, HBx activated mTOR complex 2, which can prevent YAP degradation. mTOR complex 2-mediated upregulation of YAP was observed in human HCC specimens and mouse HCC models and could be targeted with mTOR kinase inhibitors. Thus, HBV interaction with host proteins rewires HCC signaling rather than directly activating mitogenic pathways, providing an alternative paradigm for the cellular effects of a tumor-promoting virus. Significance: Integrative proteomic and genomic analysis of HBV/host interactions illuminated modifiers of hepatocellular carcinoma behavior and key signaling mechanisms in advanced disease, which suggested that HBV may have therapeutically actionable effects.
Collapse
Affiliation(s)
- Rigney E Turnham
- Division of Hematology/Oncology, University of California, San Francisco CA
- Quantitative Biosciences Institute, University of California, San Francisco CA
| | - Adriana Pitea
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Gwendolyn M Jang
- Quantitative Biosciences Institute, University of California, San Francisco CA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA
| | - Zhong Xu
- Department of Bioengineering, University of California, San Francisco CA
| | - Huat Chye Lim
- Division of Hematology/Oncology, University of California, San Francisco CA
- Quantitative Biosciences Institute, University of California, San Francisco CA
| | - Alex L Choi
- Division of Hematology/Oncology, University of California, San Francisco CA
- Quantitative Biosciences Institute, University of California, San Francisco CA
| | - John Von Dollen
- Quantitative Biosciences Institute, University of California, San Francisco CA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA
| | - Rebecca S. Levin
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA
| | - James T Webber
- Department of Bioengineering, University of California, San Francisco CA
| | - Elizabeth McCarthy
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA
| | - Junjie Hu
- Department of Bioengineering, University of California, San Francisco CA
| | - Xiaolei Li
- Department of Bioengineering, University of California, San Francisco CA
| | - Li Che
- Department of Bioengineering, University of California, San Francisco CA
| | - Ananya Singh
- Division of Hematology/Oncology, University of California, San Francisco CA
- Quantitative Biosciences Institute, University of California, San Francisco CA
| | - Alex Yoon
- Division of Hematology/Oncology, University of California, San Francisco CA
- Quantitative Biosciences Institute, University of California, San Francisco CA
| | - Gary Chan
- Division of Hematology/Oncology, University of California, San Francisco CA
- Quantitative Biosciences Institute, University of California, San Francisco CA
| | - Robin K Kelley
- Division of Hematology/Oncology, University of California, San Francisco CA
| | - Danielle L Swaney
- Quantitative Biosciences Institute, University of California, San Francisco CA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA
| | - Wei Zhang
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | | | - Fabian J Theis
- TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Manon Eckhardt
- Quantitative Biosciences Institute, University of California, San Francisco CA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, CA
| | - Xin Chen
- Department of Bioengineering, University of California, San Francisco CA
| | - Kevan M Shokat
- Quantitative Biosciences Institute, University of California, San Francisco CA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA
- Howard Hughes Medical Institute, University of California, San Francisco CA
| | - Trey Ideker
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Nevan J Krogan
- Quantitative Biosciences Institute, University of California, San Francisco CA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, CA
| | - John D Gordan
- Division of Hematology/Oncology, University of California, San Francisco CA
- Quantitative Biosciences Institute, University of California, San Francisco CA
| |
Collapse
|
7
|
Kim H, Takegahara N, Choi Y. Protocadherin-7 Regulates Monocyte Migration Through Regulation of Small GTPase RhoA and Rac1. Int J Mol Sci 2025; 26:572. [PMID: 39859288 PMCID: PMC11766416 DOI: 10.3390/ijms26020572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/30/2025] Open
Abstract
Protocadherin-7 (Pcdh7) is a member of the non-clustered protocadherin δ1 subgroup within the cadherin superfamily. Pcdh7 has been shown to control osteoclast differentiation via the protein phosphatase 2A (PP2A)-glycogen synthase kinase-3β (GSK3β)-small GTPase signaling axis. As protocadherins serve multiple biological functions, a deeper understanding of Pcdh7's biological features is valuable. Using an in vitro mouse monocyte cell culture system, we demonstrate that Pcdh7 plays a role in regulating monocyte migration by modulating the small GTPases RhoA and Rac1. Pcdh7-deficient (Pcdh7-/-) bone marrow-derived monocytes exhibited impaired migration along with the reduced activation of RhoA and Rac1. This impaired migration was rescued by transduction with constitutively active forms of RhoA and Rac1. Treatment with the PP2A-specific activator DT-061 enhanced cell migration, whereas treatment with the GSK3β-specific inhibitor AR-A014418 inhibited migration in wild-type monocytes. In contrast, treatment with DT-061 failed to restore the impaired migration in Pcdh7-/- monocytes. These findings suggest the involvement of PP2A and GSK3β in monocyte migration, although the forced activation of PP2A alone is insufficient to restore impaired migration in Pcdh7-/- monocytes. Taken together, these results indicate that Pcdh7 regulates monocyte migration through the activation of RhoA and Rac1. Given the pivotal role of cell migration in both physiological and pathological processes, our findings provide a foundation for future research into therapeutic strategies targeting Pcdh7-regulated migration.
Collapse
Affiliation(s)
| | | | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; (H.K.); (N.T.)
| |
Collapse
|
8
|
Germini D, Farhat R, Dadon L, Lacroix A, Nemati F, Rebollo A, Decaudin D, Wiels J, Brenner C. A translational study for biomarker identification of PEP-010, a pro-apoptotic peptide restoring apoptosis in cancer models. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167492. [PMID: 39218274 DOI: 10.1016/j.bbadis.2024.167492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/31/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Affiliation(s)
- D Germini
- PEP-Therapy, 111 Avenue de France, 75013 Paris, France.
| | - R Farhat
- PEP-Therapy, 111 Avenue de France, 75013 Paris, France
| | - L Dadon
- PEP-Therapy, 111 Avenue de France, 75013 Paris, France
| | - A Lacroix
- PEP-Therapy, 111 Avenue de France, 75013 Paris, France
| | - F Nemati
- Translational Research Department, Laboratory of Preclinical Investigation, PSL University, Institut Curie, 26 rue d'Ulm, Paris 75248, France
| | - A Rebollo
- Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), INSERM U1267 - CNRS UMR8258, Université de Paris, Faculté de Pharmacie, France
| | - D Decaudin
- Translational Research Department, Laboratory of Preclinical Investigation, PSL University, Institut Curie, 26 rue d'Ulm, Paris 75248, France; Department of Medical Oncology, Institut Curie, 26 rue d'Ulm, Paris 75248, France
| | - J Wiels
- Université Paris-Saclay, CNRS, Institut Gustave Roussy, Aspects métaboliques et systémiques de l'oncogénèse pour de nouvelles approches thérapeutiques, 94805 Villejuif, France
| | - C Brenner
- Université Paris-Saclay, CNRS, Institut Gustave Roussy, Aspects métaboliques et systémiques de l'oncogénèse pour de nouvelles approches thérapeutiques, 94805 Villejuif, France.
| |
Collapse
|
9
|
Shinzawa Y, Hara D, Shinguryo Y, Yokoyama S, Kawada M, Hayakawa Y. PP2A negatively regulates NK cell T-bet expression and anti-tumor effector function. Int Immunol 2024; 37:97-107. [PMID: 39404747 DOI: 10.1093/intimm/dxae057] [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: 05/16/2024] [Accepted: 10/11/2024] [Indexed: 12/28/2024] Open
Abstract
The transcription factor T-bet is essential for the anti-tumor effector function of natural killer (NK) cells, but the mechanism regulating its expression in NK cells remains unclear. In this study, we aimed to identify an NK cell-intrinsic regulator that controls T-bet expression. Using T-bet-luciferase reporter assay screening, we identified a protein phosphatase inhibitor as a potential activator of T-bet expression. A series of protein phosphatase 2A (PP2A)-specific inhibitors (PP2Ai) or PP2A siRNA induced the expression of T-bet. In PP2Ai-treated mice, the expression of T-bet and its downstream effector molecules, granzyme B and IFN-γ, was also upregulated in NK cells. Mechanistically, PP2Ai increased the phosphorylation of mTOR and ribosomal protein S6 in NK cells, and mTOR inhibitor canceled the effects of PP2Ai in NK cells. Importantly, NK cells isolated from PP2Ai-treated mice showed higher cytotoxicity and IFN-γ production; therefore, they increased the anti-tumor effector function of NK cells. Accordingly, PP2Ai treatment inhibited lung metastasis of B16 melanoma by NK cell- and mTOR-dependent mechanisms. These results suggest that PP2A negatively regulates NK cell T-bet expression and effector function by an mTOR-dependent mechanism.
Collapse
Affiliation(s)
- Yui Shinzawa
- Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Daisuke Hara
- Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Yuki Shinguryo
- Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Satoru Yokoyama
- Department of Cancer Cell Biology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Manabu Kawada
- Laboratory of Oncology, Institute of Microbial Chemistry, Tokyo, Japan
| | - Yoshihiro Hayakawa
- Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
10
|
Wu Q, Nandi D, Sharma D. TRIM-endous functional network of tripartite motif 29 (TRIM29) in cancer progression and beyond. Cancer Metastasis Rev 2024; 44:16. [PMID: 39644332 PMCID: PMC11625080 DOI: 10.1007/s10555-024-10226-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/16/2024] [Indexed: 12/09/2024]
Abstract
While most Tripartite motif (TRIM) family proteins are E3 ubiquitin ligases, some members have functions beyond the regulation of ubiquitination, impacting normal physiological processes and disease progression. TRIM29, an important member of the TRIM family, exerts a predominant influence on cancer growth, epithelial-to-mesenchymal transition, stemness and metastatic progression by directly potentiating multiple canonical oncogenic pathways. The cancer-promoting effect of TRIM29 is also evident in metabolic interventions and interference with the efficacy of cancer therapeutics. As expected for any key node in cancer, the expression of TRIM29 is tightly regulated by non-coding RNAs, epigenetic modulation, and post-translational regulation. A systematic discussion of how TRIM29 is regulated in cancer, its influences on cancer progression, and its impact on cancer therapeutics is presented in this review. We also explore the context-dependent alterations between TRIM29 function from oncogenic to tumor suppression. As TRIM29 is involved in multiple aspects of cancer progression, a better understanding of its biological impact in cancer may help improve prognosis and develop novel therapeutic combinations, leading to improved personalized cancer care.
Collapse
Affiliation(s)
- Qitong Wu
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Deeptashree Nandi
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Dipali Sharma
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA.
| |
Collapse
|
11
|
Rajput S, Jain S, Dash D, Gupta N, Rajpoot R, Upadhyaya CP, Khan ML, Koiri RK. Role of cyanotoxins in the development and promotion of cancer. Toxicol Rep 2024; 13:101798. [PMID: 39606777 PMCID: PMC11600067 DOI: 10.1016/j.toxrep.2024.101798] [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: 07/05/2024] [Revised: 10/23/2024] [Accepted: 11/02/2024] [Indexed: 11/29/2024] Open
Abstract
Cyanotoxins are primarily produced by different species of cyanobacteria, also known as blue-green algae, and have appeared to be environmental poisons that have various toxic effects on animal health, including humans. Cyanotoxins have been linked to the development and promotion of multiple cancers in recent studies. Important cyanotoxins, such as microcystins, nodularins, and cylindrospermopsin, have been found to play significant roles in developing and promoting various cancers. These toxins are generally responsible for oxidative stress, DNA damage, and disrupt cellular signaling pathways thus the development of cancers in various cells. Cancer is a multistep process caused by multiple mutations in normal cells. Microcystin-LR inhibits protein phosphatases (PP1 and PP2A), which leads to abnormal cell proliferation and tumor development. Similar inhibition of PP1 and PP2A is shown by nodularin, and in fact, their mechanism of carcinogenesis is the same as that of microcystins to some extent. Cylindrospermopsin inhibits protein synthesis and thus has genotoxic effects and may promote the development of cancer. Anatoxin-a and saxitoxins are well-known neurotoxins but, are thought to have indirect carcinogenic effects based on the fact that they can induce oxidative stress and DNA damage in cells by producing reactive oxygen species, thus further studies are needed to fully elucidate their role in the development and promotion of cancer. This review provides a detailed account of how different cyanotoxins play a role in the development and promotion of cancer.
Collapse
Affiliation(s)
- Siddharth Rajput
- Biochemistry Laboratory, Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh 470003, India
| | - Shruti Jain
- Biochemistry Laboratory, Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh 470003, India
| | - Debabrata Dash
- Biochemistry Laboratory, Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh 470003, India
| | - Nidhi Gupta
- Biochemistry Laboratory, Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh 470003, India
| | - Roshni Rajpoot
- Biochemistry Laboratory, Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh 470003, India
| | - Chandrama Prakash Upadhyaya
- Department of Biotechnology, School of Biological Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh 470003, India
| | - Mohammed Latif Khan
- Department of Botany, School of Biological Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh 470003, India
| | - Raj Kumar Koiri
- Biochemistry Laboratory, Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh 470003, India
| |
Collapse
|
12
|
Nazam N, Bownes LV, Julson JR, Quinn CH, Erwin MH, Marayati R, Markert HR, Shirley S, Stewart JE, Yoon KJ, Aye J, Ohlmeyer M, Beierle EA. Novel PP2A-Activating Compounds in Neuroblastoma. Cancers (Basel) 2024; 16:3836. [PMID: 39594793 PMCID: PMC11592631 DOI: 10.3390/cancers16223836] [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: 09/17/2024] [Revised: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Neuroblastoma (NB) remains one of the deadliest pediatric solid tumors. Recent advancements aimed at improving outcomes have been insufficient, and patients with high-risk NB continue to have a poor prognosis. Protein phosphatase 2A (PP2A) is a tumor suppressor protein downregulated in many cancers, including NB. PP2A activation has been shown to affect the malignant phenotype in other solid tumors. The present studies aim to investigate the effects of two novel PP2A activators as a NB therapeutic. METHODS Four established NB cell lines and a patient-derived xenoline were utilized to study the effect on cell viability, proliferation, motility, and in vivo tumor growth using two novel tricyclic sulfonamide PP2A activators, ATUX-3364 and ATUX-8385. RESULTS ATUX-3364 and ATUX-8385 increased PP2A activity. These PP2A activators led to decreased viability, proliferation, and motility of NB cells. Treatment of animals bearing NB tumors with ATUX-3364 or ATUX-8385 resulted in decreased tumor growth in MYCN-amplified SK-N-BE(2) tumors. At the molecular level, PP2A-based reactivation led to dephosphorylation of MYCN-S62 and decreased MYCN protein expression. CONCLUSIONS PP2A activators decreased NB cell viability, proliferation, and motility. In vivo experiments show that PP2A activators have more significant effects on tumorigenesis in MYCN-amplified tumors. Finally, phosphorylation of MYCN protein was decreased following treatment with novel sulfonamide PP2A activators. These data and mechanistic insights may be useful for developing new PP2A-based therapies that target MYCN for the treatment of NB.
Collapse
Affiliation(s)
- Nazia Nazam
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (N.N.); (L.V.B.); (J.R.J.); (C.H.Q.); (M.H.E.); (R.M.); (H.R.M.); (S.S.); (J.E.S.)
| | - Laura V. Bownes
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (N.N.); (L.V.B.); (J.R.J.); (C.H.Q.); (M.H.E.); (R.M.); (H.R.M.); (S.S.); (J.E.S.)
| | - Janet R. Julson
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (N.N.); (L.V.B.); (J.R.J.); (C.H.Q.); (M.H.E.); (R.M.); (H.R.M.); (S.S.); (J.E.S.)
| | - Colin H. Quinn
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (N.N.); (L.V.B.); (J.R.J.); (C.H.Q.); (M.H.E.); (R.M.); (H.R.M.); (S.S.); (J.E.S.)
| | - Michael H. Erwin
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (N.N.); (L.V.B.); (J.R.J.); (C.H.Q.); (M.H.E.); (R.M.); (H.R.M.); (S.S.); (J.E.S.)
| | - Raoud Marayati
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (N.N.); (L.V.B.); (J.R.J.); (C.H.Q.); (M.H.E.); (R.M.); (H.R.M.); (S.S.); (J.E.S.)
| | - Hooper R. Markert
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (N.N.); (L.V.B.); (J.R.J.); (C.H.Q.); (M.H.E.); (R.M.); (H.R.M.); (S.S.); (J.E.S.)
| | - Sorina Shirley
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (N.N.); (L.V.B.); (J.R.J.); (C.H.Q.); (M.H.E.); (R.M.); (H.R.M.); (S.S.); (J.E.S.)
| | - Jerry E. Stewart
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (N.N.); (L.V.B.); (J.R.J.); (C.H.Q.); (M.H.E.); (R.M.); (H.R.M.); (S.S.); (J.E.S.)
| | - Karina J. Yoon
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Jamie Aye
- Division of Hematology/Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | | | - Elizabeth A. Beierle
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (N.N.); (L.V.B.); (J.R.J.); (C.H.Q.); (M.H.E.); (R.M.); (H.R.M.); (S.S.); (J.E.S.)
| |
Collapse
|
13
|
Williams KB, Larsson AT, Keller BJ, Chaney KE, Williams RL, Bhunia MM, Draper GM, Jubenville TA, Hudson WA, Moertel CL, Ratner N, Largaespada DA. Pharmacogenomic synthetic lethal screens reveal hidden vulnerabilities and new therapeutic approaches for treatment of NF1-associated tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.25.585959. [PMID: 38585724 PMCID: PMC10996510 DOI: 10.1101/2024.03.25.585959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Neurofibromatosis Type 1 (NF1) is a common cancer predisposition syndrome, caused by heterozygous loss of function mutations in the tumor suppressor gene NF1. Individuals with NF1 develop benign tumors of the peripheral nervous system (neurofibromas), originating from the Schwann cell linage after somatic loss of the wild type NF1 allele, some of which progress further to malignant peripheral nerve sheath tumors (MPNST). There is only one FDA approved targeted therapy for symptomatic plexiform neurofibromas and none approved for MPNST. The genetic basis of NF1 syndrome makes associated tumors ideal for using synthetic drug sensitivity approaches to uncover therapeutic vulnerabilities. We developed a drug discovery pipeline to identify therapeutics for NF1-related tumors using isogeneic pairs of NF1-proficient and deficient immortalized human Schwann cells. We utilized these in a large-scale high throughput screen (HTS) for drugs that preferentially kill NF1-deficient cells, through which we identified 23 compounds capable of killing NF1-deficient Schwann cells with selectivity. Multiple hits from this screen clustered into classes defined by method of action. Four clinically interesting drugs from these classes were tested in vivo using both a genetically engineered mouse model of high-grade peripheral nerve sheath tumors and human MPNST xenografts. All drugs tested showed single agent efficacy in these models as well as significant synergy when used in combination with the MEK inhibitor Selumetinib. This HTS platform yielded novel therapeutically relevant compounds for the treatment of NF1-associated tumors and can serve as a tool to rapidly evaluate new compounds and combinations in the future.
Collapse
Affiliation(s)
- Kyle B Williams
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alex T Larsson
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Bryant J Keller
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Katherine E Chaney
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229-0713, USA
| | - Rory L Williams
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Minu M Bhunia
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Twin Cities, Minneapolis, Minnesota 55455, USA
| | - Garrett M Draper
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Tyler A Jubenville
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Wendy A Hudson
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christopher L Moertel
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229-0713, USA
| | - David A Largaespada
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Twin Cities, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
14
|
Niu MY, Liu YJ, Shi JJ, Chen RY, Zhang S, Li CY, Cao JF, Yang GJ, Chen J. The Emerging Role of Ubiquitin-Specific Protease 36 (USP36) in Cancer and Beyond. Biomolecules 2024; 14:572. [PMID: 38785979 PMCID: PMC11118191 DOI: 10.3390/biom14050572] [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: 03/28/2024] [Revised: 05/07/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
The balance between ubiquitination and deubiquitination is instrumental in the regulation of protein stability and maintenance of cellular homeostasis. The deubiquitinating enzyme, ubiquitin-specific protease 36 (USP36), a member of the USP family, plays a crucial role in this dynamic equilibrium by hydrolyzing and removing ubiquitin chains from target proteins and facilitating their proteasome-dependent degradation. The multifaceted functions of USP36 have been implicated in various disease processes, including cancer, infections, and inflammation, via the modulation of numerous cellular events, including gene transcription regulation, cell cycle regulation, immune responses, signal transduction, tumor growth, and inflammatory processes. The objective of this review is to provide a comprehensive summary of the current state of research on the roles of USP36 in different pathological conditions. By synthesizing the findings from previous studies, we have aimed to increase our understanding of the mechanisms underlying these diseases and identify potential therapeutic targets for their treatment.
Collapse
Affiliation(s)
- Meng-Yao Niu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China; (M.-Y.N.); (Y.-J.L.); (J.-J.S.); (R.-Y.C.); (C.-Y.L.); (J.-F.C.)
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
| | - Yan-Jun Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China; (M.-Y.N.); (Y.-J.L.); (J.-J.S.); (R.-Y.C.); (C.-Y.L.); (J.-F.C.)
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
| | - Jin-Jin Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China; (M.-Y.N.); (Y.-J.L.); (J.-J.S.); (R.-Y.C.); (C.-Y.L.); (J.-F.C.)
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
| | - Ru-Yi Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China; (M.-Y.N.); (Y.-J.L.); (J.-J.S.); (R.-Y.C.); (C.-Y.L.); (J.-F.C.)
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
| | - Shun Zhang
- Ningbo No.2 Hospital, Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China;
| | - Chang-Yun Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China; (M.-Y.N.); (Y.-J.L.); (J.-J.S.); (R.-Y.C.); (C.-Y.L.); (J.-F.C.)
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
| | - Jia-Feng Cao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China; (M.-Y.N.); (Y.-J.L.); (J.-J.S.); (R.-Y.C.); (C.-Y.L.); (J.-F.C.)
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
| | - Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China; (M.-Y.N.); (Y.-J.L.); (J.-J.S.); (R.-Y.C.); (C.-Y.L.); (J.-F.C.)
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
| | - Jiong Chen
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
- Ningbo No.2 Hospital, Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China;
| |
Collapse
|
15
|
Fan C, Yang X, Yan L, Shi Z. Oxidative stress is two-sided in the treatment of acute myeloid leukemia. Cancer Med 2024; 13:e6806. [PMID: 38715546 PMCID: PMC11077289 DOI: 10.1002/cam4.6806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/14/2023] [Accepted: 11/27/2023] [Indexed: 05/12/2024] Open
Abstract
INTRODUCTION Oxidative stress caused by elevated ROS, as a novel therapeutic mechanism, has been implicated in various tumors including AML. AML cells are chronically under oxidative stress, yet overreliance on ROS production makes tumor cells increasingly vulnerable to further damage. Reducing the cytotoxic effect of ROS on normal cells while killing leukemia stem cell (LSC) with high levels of reactive oxygen species is a new challenge for oxidative stress therapy in leukemia. METHODS By searching literature databases, we summarized recent relevant studies. The relationship of ROS on AML genes, signaling pathways, and transcription factors, and the correlation of ROS with AML bone marrow microenvironment and autophagy were summarized. In addition, we summarize the current status of research on ROS and AML therapeutics. Finally, we discuss the research progress on redox resistance in AML. RESULTS This review discusses the evidence showing the link between redox reactions and the progression of AML and compiles the latest research findings that will facilitate future biological studies of redox effects associated with AML treatment. CONCLUSION We believe that exploiting this unique oxidative stress property of AML cells may provide a new way to prevent relapse and drug resistance.
Collapse
Affiliation(s)
- Chenyang Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Xiangdong Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Lixiang Yan
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Zhexin Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| |
Collapse
|
16
|
Ramachandran D, Tyrer JP, Kommoss S, DeFazio A, Riggan MJ, Webb PM, Fasching PA, Lambrechts D, García MJ, Rodríguez-Antona C, Goodman MT, Modugno F, Moysich KB, Karlan BY, Lester J, Kjaer SK, Jensen A, Høgdall E, Goode EL, Cliby WA, Kumar A, Wang C, Cunningham JM, Winham SJ, Monteiro AN, Schildkraut JM, Cramer DW, Terry KL, Titus L, Bjorge L, Thomsen LCV, Pejovic T, Høgdall CK, McNeish IA, May T, Huntsman DG, Pfisterer J, Canzler U, Park-Simon TW, Schröder W, Belau A, Hanker L, Harter P, Sehouli J, Kimmig R, de Gregorio N, Schmalfeldt B, Baumann K, Hilpert F, Burges A, Winterhoff B, Schürmann P, Speith LM, Hillemanns P, Berchuck A, Johnatty SE, Ramus SJ, Chenevix-Trench G, Pharoah PDP, Dörk T, Heitz F. Genome-wide association analyses of ovarian cancer patients undergoing primary debulking surgery identify candidate genes for residual disease. NPJ Genom Med 2024; 9:19. [PMID: 38443389 PMCID: PMC10915171 DOI: 10.1038/s41525-024-00395-y] [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: 06/14/2023] [Accepted: 01/15/2024] [Indexed: 03/07/2024] Open
Abstract
Survival from ovarian cancer depends on the resection status after primary surgery. We performed genome-wide association analyses for resection status of 7705 ovarian cancer patients, including 4954 with high-grade serous carcinoma (HGSOC), to identify variants associated with residual disease. The most significant association with resection status was observed for rs72845444, upstream of MGMT, in HGSOC (p = 3.9 × 10-8). In gene-based analyses, PPP2R5C was the most strongly associated gene in HGSOC after stage adjustment. In an independent set of 378 ovarian tumours from the AGO-OVAR 11 study, variants near MGMT and PPP2R5C correlated with methylation and transcript levels, and PPP2R5C mRNA levels predicted progression-free survival in patients with residual disease. MGMT encodes a DNA repair enzyme, and PPP2R5C encodes the B56γ subunit of the PP2A tumour suppressor. Our results link heritable variation at these two loci with resection status in HGSOC.
Collapse
Grants
- R21 CA267050 NCI NIH HHS
- K05 CA154337 NCI NIH HHS
- UL1 TR000124 NCATS NIH HHS
- P50 CA105009 NCI NIH HHS
- K07 CA080668 NCI NIH HHS
- P30 CA076292 NCI NIH HHS
- R01 CA076016 NCI NIH HHS
- R01 CA248288 NCI NIH HHS
- U19 CA148112 NCI NIH HHS
- R01 CA149429 NCI NIH HHS
- Wellcome Trust
- UL1 TR001881 NCATS NIH HHS
- P50 CA136393 NCI NIH HHS
- M01 RR000056 NCRR NIH HHS
- R01 CA095023 NCI NIH HHS
- P30 CA047904 NCI NIH HHS
- R01 CA058598 NCI NIH HHS
- R01 CA054419 NCI NIH HHS
- P30 CA015083 NCI NIH HHS
- Deutsche Forschungsgemeinschaft (German Research Foundation)
- The Ovarian Cancer Association Consortium is funded by generous contributions from its research investigators and through anonymous donations. OCAC was funded by a grant from the Ovarian Cancer Research Fund (OCRF). The OCAC OncoArray genotyping project was funded through grants from the U.S. National Institutes of Health (CA1X01HG007491-01 (C.I.A.), U19-CA148112 (T.A.S.), R01-CA149429 (C.M.P.) and R01-CA058598 (M.T.G.); Canadian Institutes of Health Research (MOP-86727 (L.E.K.) and the Ovarian Cancer Research Fund (A.B.). The COGS project was funded through a European Commission’s Seventh Framework Programme grant (agreement number 223175 - HEALTH-F2-2009-223175) and in part by the US National Cancer Institute GAME-ON Post-GWAS Initiative (U19-CA148112). This study made use of data generated by the Wellcome Trust Case Control consortium that was funded by the Wellcome Trust under award 076113. The results published are in part based upon data generated by The Cancer Genome Atlas Pilot Project established by the National Cancer Institute and National Human Genome Research Institute (dbGap accession number phs000178.v8.p7). Funding for individual studies: AUS: The Australian Ovarian Cancer Study (AOCS) was supported by the U.S. Army Medical Research and Materiel Command (DAMD17-01-1-0729), National Health & Medical Research Council of Australia (199600, 400413 and 400281), Cancer Councils of New South Wales, Victoria, Queensland, South Australia and Tasmania and Cancer Foundation of Western Australia (Multi-State Applications 191, 211 and 182). AOCS gratefully acknowledges additional support from Ovarian Cancer Australia and the Peter MacCallum Foundation; BAV: ELAN Funds of the University of Erlangen-Nuremberg; BEL: National Kankerplan; CNI: Instituto de Salud Carlos III (PI 19/01730); Ministerio de Economía y Competitividad (SAF2012); HAW: U.S. National Institutes of Health (R01-CA58598, N01-CN-55424 and N01-PC-67001); HOP: University of Pittsburgh School of Medicine Dean’s Faculty Advancement Award (F. Modugno), Department of Defense (DAMD17-02-1-0669, OC20085) and United States National Cancer Institute (R21-CA267050, K07-CA080668, R01-CA95023, MO1-RR000056); LAX: American Cancer Society Early Detection Professorship (SIOP-06-258-01-COUN) and the National Center for Advancing Translational Sciences (NCATS), Grant UL1TR000124; MAC: National Institutes of Health (R01-CA2482288, P30-CA15083, P50-CA136393); Mayo Foundation; Minnesota Ovarian Cancer Alliance; Fred C. and Katherine B. Andersen Foundation; Fraternal Order of Eagles; MAL: Funding for this study was provided by research grant R01- CA61107 from the National Cancer Institute, Bethesda, MD, research grant 94 222 52 from the Danish Cancer Society, Copenhagen, Denmark, the Mermaid I project; and the Mermaid III project; MAY: National Institutes of Health (R01-CA2482288, P30-CA15083, P50-CA136393); Mayo Foundation; Minnesota Ovarian Cancer Alliance; Fred C. and Katherine B. Andersen Foundation; MOF: Moffitt Cancer Center, Merck Pharmaceuticals, the state of Florida, Hillsborough County, and the city of Tampa; NCO: National Institutes of Health (R01-CA76016) and the Department of Defense (DAMD17-02-1-0666); NEC: National Institutes of Health R01-CA54419 and P50-CA105009 and Department of Defense W81XWH-10-1-02802; NOR: Helse Vest, The Norwegian Cancer Society, The Research Council of Norway; OPL: National Health and Medical Research Council (NHMRC) of Australia (APP1025142, APP1120431) and Brisbane Women’s Club; ORE: Sherie Hildreth Ovarian Cancer (SHOC) Foundation; PVD: Canadian Cancer Society and Cancer Research Society GRePEC Program; SRO: Cancer Research UK (C536/A13086, C536/A6689) and Imperial Experimental Cancer Research Centre (C1312/A15589); UHN: Princess Margaret Cancer Centre Foundation-Bridge for the Cure; VAN: BC Cancer Foundation, VGH & UBC Hospital Foundation; VTL: NIH K05-CA154337; WMH: National Health and Medical Research Council of Australia, Enabling Grants ID 310670 & ID 628903. Cancer Institute NSW Grants 12/RIG/1-17 & 15/RIG/1-16. The AGO-OVAR 11 study was funded by Roche Pharma AG.
- National Health and Medical Research Council (NHMRC) of Australia (APP1025142, APP1120431) and Brisbane Women’s Club
Collapse
Affiliation(s)
| | - Jonathan P Tyrer
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Stefan Kommoss
- Department of Women's Health, Tuebingen University Hospital, Tuebingen, Germany
| | - Anna DeFazio
- Centre for Cancer Research, The Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, Australia
- Discipline of Obstetrics and Gynaecology, The University of Sydney, Sydney, NSW, Australia
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia
- The Daffodil Centre, The University of Sydney, a joint venture with Cancer Council NSW, Sydney, NSW, Australia
| | - Marjorie J Riggan
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Duke University Medical Center, Durham, NC, USA
| | - Penelope M Webb
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Peter A Fasching
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - María J García
- Biochemistry and Molecular Biology area, Department of Basic Health Sciences, Faculty of Health Sciences, Rey Juan Carlos University, Madrid, Spain
| | - Cristina Rodríguez-Antona
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Marc T Goodman
- Cancer Prevention and Control Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Francesmary Modugno
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Women's Cancer Research Center, Magee-Womens Research Institute and Hillman Cancer Center, Pittsburgh, PA, USA
| | - Kirsten B Moysich
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Beth Y Karlan
- David Geffen School of Medicine, Department of Obstetrics and Gynecology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Jenny Lester
- David Geffen School of Medicine, Department of Obstetrics and Gynecology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Susanne K Kjaer
- Department of Virus, Lifestyle and Genes, Danish Cancer Institute, Copenhagen, Denmark
- Department of Gynaecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Allan Jensen
- Department of Virus, Lifestyle and Genes, Danish Cancer Institute, Copenhagen, Denmark
| | - Estrid Høgdall
- Department of Pathology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ellen L Goode
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - William A Cliby
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Mayo Clinic, Rochester, MN, USA
| | - Amanika Kumar
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Mayo Clinic, Rochester, MN, USA
| | - Chen Wang
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, MN, USA
| | - Julie M Cunningham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Stacey J Winham
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, MN, USA
| | - Alvaro N Monteiro
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Joellen M Schildkraut
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Daniel W Cramer
- Obstetrics and Gynecology Epidemiology Center, Department of Obstetrics and Gyneclogy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kathryn L Terry
- Obstetrics and Gynecology Epidemiology Center, Department of Obstetrics and Gyneclogy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Linda Titus
- Norris Cotton Cancer Center, Lebanon, NH, USA
| | - Line Bjorge
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Liv Cecilie Vestrheim Thomsen
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Tanja Pejovic
- Department of ObGyn, Providence Medical Center, Medford, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Claus K Høgdall
- Department of Gynaecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Iain A McNeish
- Division of Cancer and Ovarian Cancer Action Research Centre, Department Surgery & Cancer, Imperial College London, London, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Taymaa May
- Division of Gynecologic Oncology, University Health Network, Princess Margaret Hospital, Toronto, ON, Canada
| | - David G Huntsman
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, BC, Canada
| | | | - Ulrich Canzler
- University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| | | | - Willibald Schröder
- Klinikum Bremen-Mitte, Bremen, Germany
- Gynaekologicum Bremen, Bremen, Germany
| | - Antje Belau
- University Hospital Greifswald, Greifswald, Germany
- Frauenarztpraxis Belau, Greifswald, Germany
| | - Lars Hanker
- University Hospital Frankfurt, Frankfurt, Germany
- University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Philipp Harter
- Department of Gynecology and Gynecologic Oncology, Evangelische Kliniken Essen-Mitte (KEM), Essen, Germany
| | - Jalid Sehouli
- Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Rainer Kimmig
- University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Nikolaus de Gregorio
- University Hospital Ulm, Ulm, Germany
- SLK-Kliniken Heilbronn, Klinikum am Gesundbrunnen, Heilbronn, Germany
| | | | - Klaus Baumann
- University Hospital Gießen and Marburg, Site Marburg, Marburg, Germany
- Klinikum Ludwigshafen, Ludwigshafen, Germany
| | - Felix Hilpert
- University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
- Krankenhaus Jerusalem, Mammazentrum Hamburg, Hamburg, Germany
| | | | - Boris Winterhoff
- Department of Obstetrics, Gynecology and Women's Health, Division of Gynecologic Oncology, University of Minnesota, Minneapolis, MN, USA
| | - Peter Schürmann
- Gynaecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Lisa-Marie Speith
- Gynaecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Peter Hillemanns
- Gynaecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Andrew Berchuck
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Duke University Medical Center, Durham, NC, USA
| | - Sharon E Johnatty
- Cancer Division, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Susan J Ramus
- School of Clinical Medicine, UNSW Medicine and Health, University of NSW Sydney, Sydney, NSW, Australia
- Adult Cancer Program, Lowy Cancer Research Centre, University of NSW Sydney, Sydney, NSW, Australia
| | | | - Paul D P Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, West Hollywood, CA, USA
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Thilo Dörk
- Gynaecology Research Unit, Hannover Medical School, Hannover, Germany.
| | - Florian Heitz
- Department of Gynecology and Gynecologic Oncology, Evangelische Kliniken Essen-Mitte (KEM), Essen, Germany.
- Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany.
- Department of Gynecology and Gynecological Oncology, HSK, Dr. Horst-Schmidt Klinik, Wiesbaden, Wiesbaden, Germany.
| |
Collapse
|
17
|
Zhang Q, Yuan Y, Cao S, Kang N, Qiu F. Withanolides: Promising candidates for cancer therapy. Phytother Res 2024; 38:1104-1158. [PMID: 38176694 DOI: 10.1002/ptr.8090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/14/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024]
Abstract
Natural products have played a significant role throughout history in the prevention and treatment of numerous diseases, particularly cancers. As a natural product primarily derived from various medicinal plants in the Withania genus, withanolides have been shown in several studies to exhibit potential activities in cancer treatment. Consequently, understanding the molecular mechanism of withanolides could herald the discovery of new anticancer agents. Withanolides have been studied widely, especially in the last 20 years, and attracted the attention of numerous researchers. Currently, over 1200 withanolides have been classified, with approximately a quarter of them having been reported in the literature to be able to modulate the survival and death of cancer cells through multiple avenues. To what extent, though, has the anticancer effects of these compounds been studied? How far are they from being developed into clinical drugs? What are their potential, characteristic features, and challenges? In this review, we elaborate on the current knowledge of natural compounds belonging to this class and provide an overview of their natural sources, anticancer activity, mechanism of action, molecular targets, and implications for anticancer drug research. In addition, direct targets and clinical research to guide the design and implementation of future preclinical and clinical studies to accelerate the application of withanolides have been highlighted.
Collapse
Affiliation(s)
- Qiang Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - YongKang Yuan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Shijie Cao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Ning Kang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Feng Qiu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| |
Collapse
|
18
|
Johnson H, Narayan S, Sharma AK. Altering phosphorylation in cancer through PP2A modifiers. Cancer Cell Int 2024; 24:11. [PMID: 38184584 PMCID: PMC10770906 DOI: 10.1186/s12935-023-03193-1] [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: 09/24/2023] [Accepted: 12/25/2023] [Indexed: 01/08/2024] Open
Abstract
Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase integral to the regulation of many cellular processes. Due to the deregulation of PP2A in cancer, many of these processes are turned toward promoting tumor progression. Considerable research has been undertaken to discover molecules capable of modulating PP2A activity in cancer. Because PP2A is capable of immense substrate specificity across many cellular processes, the therapeutic targeting of PP2A in cancer can be completed through either enzyme inhibitors or activators. PP2A modulators likewise tend to be effective in drug-resistant cancers and work synergistically with other known cancer therapeutics. In this review, we will discuss the patterns of PP2A deregulation in cancer, and its known downstream signaling pathways important for cancer regulation, along with many activators and inhibitors of PP2A known to inhibit cancer progression.
Collapse
Affiliation(s)
- Hannah Johnson
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Satya Narayan
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL, 32610, USA
| | - Arun K Sharma
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
19
|
Zhu P, Wu X, Ni L, Chen K, Dong Z, Du J, Kong F, Mao Y, Tao H, Chu M, Mao H, Yang H, Liu Q, Gan M, Geng D. Inhibition of PP2A ameliorates intervertebral disc degeneration by reducing annulus fibrosus cells apoptosis via p38/MAPK signal pathway. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166888. [PMID: 37722489 DOI: 10.1016/j.bbadis.2023.166888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/05/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023]
Abstract
BACKGROUND Intervertebral disc degeneration (IVDD) is considered one of the main reasons for low back pain (LBP). To date, the specific pathology of IVDD remains unclear. The annulus fibrosus (AF) is an important part of the intervertebral disc, and AF cell oxidative stress, apoptosis plays a vital role in disc degeneration. Protein phosphatase 2 A (PP2A), a serine/threonine phosphatase, has regulatory functions in various processes, including apoptosis and autophagy. However, thus far, the effect of PP2A on IVDD is not clear. METHODS AF cells derived from caudal intervertebral discs in SD rats were used to analyze the levels of oxidative stress, apoptosis and degeneration as well as PP2A expression. A PP2A agonist (FTY720), inhibitor (microcystin-LR) and siRNA (si-PPP2CA) were employed in IVDD induced by H2O2 to investigate the levels of apoptosis and degeneration. The p38/MAPK signal pathways were evaluated, and a p38 inhibitor (SB203580) and ERK inhibitor (U0126) were added for verification. Finally, FTY720 and microcystin-LR were administered to IVDD rats to assess the effects on levels of apoptosis and degeneration and the relief of IVDD. RESULTS The expression of PP2A was increased in rat AF cells after H2O2 intervention. The levels of apoptosis and degeneration were higher with upregulation of PP2A but were significantly reduced after inhibition of PP2A. The PP2A inhibitor relieved cell apoptosis and degeneration by downregulating the p38/MAPK pathway. In vivo, the knockdown of PP2A resulted in a more complete morphology of discs and less apoptotic and degenerative expression. CONCLUSIONS This study suggests that the downregulation of PP2 A could reduce AF cell apoptosis and degeneration via the p38/MAPK pathway. It also revealed that the inhibition of PP2 A is expected to be a therapeutic target for IVDD.
Collapse
Affiliation(s)
- Pengfei Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Xiexing Wu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Li Ni
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Kai Chen
- Department of Orthopedics, Hai'an People's Hospital, Hai'an 226600, Jiangsu, China
| | - Zhongchen Dong
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Jiacheng Du
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Fanchen Kong
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yubo Mao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Huaqiang Tao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Miao Chu
- Department of Orthopedics, Yixing People's Hospital, Yixing 214200, Jiangsu, China
| | - Haiqin Mao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Qinbai Liu
- Department of Orthopaedics, Lianshui People's Hospital of Kangda College Affiliated to Nanjing Medical University, Huai'an 223001, Jiangsu, China; Department of Orthopaedics, Nanjing First Hospital, Nanjing Medical University, Nanjing 210000, Jiangsu, China.
| | - Minfeng Gan
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Dechun Geng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
20
|
Hermawan A, Wulandari F, Yudi Utomo R, Asmah Susidarti R, Kirihata M, Meiyanto E. Transcriptomics analyses reveal the effects of Pentagamaboronon-0-ol on PI3K/Akt and cell cycle of HER2+ breast cancer cells. Saudi Pharm J 2023; 31:101847. [PMID: 38028209 PMCID: PMC10652209 DOI: 10.1016/j.jsps.2023.101847] [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: 07/04/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Monoclonal antibodies and targeted therapies against HER2+ breast cancer has improved overall and disease-free survival in patients; however, encountering drug resistance causes recurrence, necessitating the development of newer HER2-targeted medications. A curcumin analog PGB-0-ol showed most cytotoxicity against HCC1954 HER2+ breast cancer cells than against other subtypes of breast cancer cells. Objective Here, we employed next-generation sequencing technology to elucidate the molecular mechanism underlying the effect of PGB-0-ol on HCC1954 HER2+ breast cancer cells. Methods The molecular mechanism underlying the action of PGB-0-ol on HCC1954 HER2+ breast cancer cells was determined using next-generation sequencing technologies. Additional bioinformatics studies were performed, including gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, disease-gene, and drug-gene associations, network topology analysis (NTA), and gene set enrichment analysis (GSEA). Results We detected 2,263 differentially expressed genes (DEGs) (1,459 upregulated and 804 downregulated) in the PGB-0-ol- and DMSO-treated HCC1954 cells. KEGG enrichment data revealed the control of phosphatidylinositol signaling system, and ErbB signaling following PGB-0-ol treatment. Gene ontology (GO) enrichment analysis demonstrated that these DEGs governed cell cycle, participated in the mitotic spindle and nuclear membrane, and controlled kinase activity at the molecular level. According to the NTA data for GO enrichment, GSEA data for KEGG, drug-gene and disease-gene, PGB-0-ol regulated PI3K/Akt signaling and cell cycle in breast cancer. Overall, our investigation revealed the transcriptomic profile of PGB-0-ol-treated HCC1954 breast cancer cells following PGB-0-ol therapy. Bioinformatics analyses showed that PI3K/Akt signaling and cell cycle was modulated. However, further studies are required to validate the findings of this study.
Collapse
Affiliation(s)
- Adam Hermawan
- Laboratory of Macromolecular Engineering, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, 55281, Yogyakarta, Indonesia
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, 55281, Yogyakarta, Indonesia
- Laboratory of Advanced Pharmaceutical Sciences. APSLC Building, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, 55281, Yogyakarta, Indonesia
| | - Febri Wulandari
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, 55281, Yogyakarta, Indonesia
| | - Rohmad Yudi Utomo
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, 55281, Yogyakarta, Indonesia
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, 55281, Yogyakarta, Indonesia
| | - Ratna Asmah Susidarti
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, 55281, Yogyakarta, Indonesia
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, 55281, Yogyakarta, Indonesia
| | - Mitsunori Kirihata
- Research Center for BNCT, Osaka Metropolitan University, 1-2, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8570, Japan
| | - Edy Meiyanto
- Laboratory of Macromolecular Engineering, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, 55281, Yogyakarta, Indonesia
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, 55281, Yogyakarta, Indonesia
| |
Collapse
|
21
|
Di Mambro A, Arroyo-Berdugo Y, Fioretti T, Randles M, Cozzuto L, Rajeeve V, Cevenini A, Austin MJ, Esposito G, Ponomarenko J, Lucas CM, Cutillas P, Gribben J, Williams O, Calle Y, Patel B, Esposito MT. SET-PP2A complex as a new therapeutic target in KMT2A (MLL) rearranged AML. Oncogene 2023; 42:3670-3683. [PMID: 37891368 PMCID: PMC10709139 DOI: 10.1038/s41388-023-02840-1] [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: 02/21/2023] [Revised: 09/07/2023] [Accepted: 09/13/2023] [Indexed: 10/29/2023]
Abstract
KMT2A-rearranged (KMT2A-R) is an aggressive and chemo-refractory acute leukemia which mostly affects children. Transcriptomics-based characterization and chemical interrogation identified kinases as key drivers of survival and drug resistance in KMT2A-R leukemia. In contrast, the contribution and regulation of phosphatases is unknown. In this study we uncover the essential role and underlying mechanisms of SET, the endogenous inhibitor of Ser/Thr phosphatase PP2A, in KMT2A-R-leukemia. Investigation of SET expression in acute myeloid leukemia (AML) samples demonstrated that SET is overexpressed, and elevated expression of SET is correlated with poor prognosis and with the expression of MEIS and HOXA genes in AML patients. Silencing SET specifically abolished the clonogenic ability of KMT2A-R leukemic cells and the transcription of KMT2A targets genes HOXA9 and HOXA10. Subsequent mechanistic investigations showed that SET interacts with both KMT2A wild type and fusion proteins, and it is recruited to the HOXA10 promoter. Pharmacological inhibition of SET by FTY720 disrupted SET-PP2A interaction leading to cell cycle arrest and increased sensitivity to chemotherapy in KMT2A-R-leukemic models. Phospho-proteomic analyses revealed that FTY720 reduced the activity of kinases regulated by PP2A, including ERK1, GSK3β, AURB and PLK1 and led to suppression of MYC, supporting the hypothesis of a feedback loop among PP2A, AURB, PLK1, MYC, and SET. Our findings illustrate that SET is a novel player in KMT2A-R leukemia and they provide evidence that SET antagonism could serve as a novel strategy to treat this aggressive leukemia.
Collapse
Affiliation(s)
| | | | - Tiziana Fioretti
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore, Napoli, Italy
| | - Michael Randles
- Chester Centre for Leukaemia Research, Chester Medical School, University of Chester, Chester, UK
| | - Luca Cozzuto
- Centre Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Armando Cevenini
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore, Napoli, Italy
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Michael J Austin
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Gabriella Esposito
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore, Napoli, Italy
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Julia Ponomarenko
- Centre Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- University Pompeu Fabra (UPF), Barcelona, Spain
| | - Claire M Lucas
- Chester Centre for Leukaemia Research, Chester Medical School, University of Chester, Chester, UK
| | - Pedro Cutillas
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - John Gribben
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Owen Williams
- Great Ormond Street Institute of Child Health London, UCL, London, UK
| | - Yolanda Calle
- School of Life and Health Sciences, University of Roehampton, London, UK
| | - Bela Patel
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Maria Teresa Esposito
- School of Life and Health Sciences, University of Roehampton, London, UK.
- School of Biosciences, University of Surrey, Guildford, UK.
| |
Collapse
|
22
|
Dow LF, Case AM, Paustian MP, Pinkerton BR, Simeon P, Trippier PC. The evolution of small molecule enzyme activators. RSC Med Chem 2023; 14:2206-2230. [PMID: 37974956 PMCID: PMC10650962 DOI: 10.1039/d3md00399j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/20/2023] [Indexed: 11/19/2023] Open
Abstract
There is a myriad of enzymes within the body responsible for maintaining homeostasis by providing the means to convert substrates to products as and when required. Physiological enzymes are tightly controlled by many signaling pathways and their products subsequently control other pathways. Traditionally, most drug discovery efforts focus on identifying enzyme inhibitors, due to upregulation being prevalent in many diseases and the existence of endogenous substrates that can be modified to afford inhibitor compounds. As enzyme downregulation and reduction of endogenous activators are observed in multiple diseases, the identification of small molecules with the ability to activate enzymes has recently entered the medicinal chemistry toolbox to afford chemical probes and potential therapeutics as an alternative means to intervene in diseases. In this review we highlight the progress made in the identification and advancement of non-kinase enzyme activators and their potential in treating various disease states.
Collapse
Affiliation(s)
- Louise F Dow
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
| | - Alfie M Case
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
| | - Megan P Paustian
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
| | - Braeden R Pinkerton
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
| | - Princess Simeon
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center Omaha NE 68106 USA
- UNMC Center for Drug Discovery, University of Nebraska Medical Center Omaha NE 68106 USA
| |
Collapse
|
23
|
Chong SJF, Zhu F, Dashevsky O, Mizuno R, Lai JX, Hackett L, Ryan CE, Collins MC, Iorgulescu JB, Guièze R, Penailillo J, Carrasco R, Hwang YC, Muñoz DP, Bouhaddou M, Lim YC, Wu CJ, Allan JN, Furman RR, Goh BC, Pervaiz S, Coppé JP, Mitsiades CS, Davids MS. Hyperphosphorylation of BCL-2 family proteins underlies functional resistance to venetoclax in lymphoid malignancies. J Clin Invest 2023; 133:e170169. [PMID: 37751299 PMCID: PMC10645378 DOI: 10.1172/jci170169] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 09/19/2023] [Indexed: 09/27/2023] Open
Abstract
The B cell leukemia/lymphoma 2 (BCL-2) inhibitor venetoclax is effective in chronic lymphocytic leukemia (CLL); however, resistance may develop over time. Other lymphoid malignancies such as diffuse large B cell lymphoma (DLBCL) are frequently intrinsically resistant to venetoclax. Although genomic resistance mechanisms such as BCL2 mutations have been described, this probably only explains a subset of resistant cases. Using 2 complementary functional precision medicine techniques - BH3 profiling and high-throughput kinase activity mapping - we found that hyperphosphorylation of BCL-2 family proteins, including antiapoptotic myeloid leukemia 1 (MCL-1) and BCL-2 and proapoptotic BCL-2 agonist of cell death (BAD) and BCL-2 associated X, apoptosis regulator (BAX), underlies functional mechanisms of both intrinsic and acquired resistance to venetoclax in CLL and DLBCL. Additionally, we provide evidence that antiapoptotic BCL-2 family protein phosphorylation altered the apoptotic protein interactome, thereby changing the profile of functional dependence on these prosurvival proteins. Targeting BCL-2 family protein phosphorylation with phosphatase-activating drugs rewired these dependencies, thus restoring sensitivity to venetoclax in a panel of venetoclax-resistant lymphoid cell lines, a resistant mouse model, and in paired patient samples before venetoclax treatment and at the time of progression.
Collapse
MESH Headings
- Mice
- Animals
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Drug Resistance, Neoplasm/genetics
- Proto-Oncogene Proteins c-bcl-2/genetics
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- bcl-X Protein/genetics
- Apoptosis Regulatory Proteins
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Cell Line, Tumor
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Apoptosis/genetics
- Myeloid Cell Leukemia Sequence 1 Protein/genetics
- Myeloid Cell Leukemia Sequence 1 Protein/metabolism
Collapse
Affiliation(s)
- Stephen Jun Fei Chong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Fen Zhu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Olga Dashevsky
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Rin Mizuno
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Jolin X.H. Lai
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Liam Hackett
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Christine E. Ryan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Mary C. Collins
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - J. Bryan Iorgulescu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Romain Guièze
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Johany Penailillo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Ruben Carrasco
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Yeonjoo C. Hwang
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| | - Denise P. Muñoz
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| | - Mehdi Bouhaddou
- Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, California, USA
| | - Yaw Chyn Lim
- Cancer Science Institute, National University of Singapore, Singapore
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - John N. Allan
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, New York, USA
| | - Richard R. Furman
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, New York, USA
| | - Boon Cher Goh
- Cancer Science Institute, National University of Singapore, Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jean-Philippe Coppé
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| | - Constantine S. Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew S. Davids
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Zhang Z, Yang Y, Xu Y, Liu Y, Li H, Chen L. Molecular targets and mechanisms of anti-cancer effects of withanolides. Chem Biol Interact 2023; 384:110698. [PMID: 37690745 DOI: 10.1016/j.cbi.2023.110698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/18/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
Withanolides are a class of natural products with a steroidal lactone structure that exhibit a broad spectrum of anti-cancer effects. To date, several studies have shown that their possible mechanisms in cancer development and progression are associated with the regulation of cell proliferation, apoptosis, metastasis, and angiogenesis. Withanolides can also attenuate inflammatory responses, as well as modulate the genomic instability and energy metabolism of cancer cells. In addition, they may improve the safety and efficacy of cancer treatments as adjuvants to traditional cancer therapeutics. Herein, we summarize the molecular targets and mechanisms of withanolides in different cancers, as well as their current clinical studies on them.
Collapse
Affiliation(s)
- Zhiruo Zhang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yueying Yang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yang Xu
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yang Liu
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hua Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; Institute of Structural Pharmacology & TCM Chemical Biology, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Lixia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
25
|
Chuan H, Li B, Wang Z, Li J, Xie P, Liu Y. Visualization Tools for Detecting Microcystin-LR in the Biological System via Near-Infrared Fluorescent Probes. Anal Chem 2023; 95:14219-14227. [PMID: 37703515 DOI: 10.1021/acs.analchem.3c01992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Numerous toxicological and epidemiological studies have shown that microcystin-LR (MC-LR) could cause a variety of toxicity to humans and animals. However, the absence of effective methods to trace MC-LR in biological systems has hindered the in-depth understanding of the mechanism of MC-LR toxicity. Near-infrared (NIR) fluorescent probes are crucial tools for accurate visualization and in-depth study of specific molecules in biological systems. Due to the lack of effective design strategies, NIR fluorescent probes for imaging MC-LR specifically in biological systems have not been reported yet. In order to address this pressing issue, herein, we have introduced a new and facile strategy to improve MC-LR detection and imaging in biological systems, and based on this design strategy, three NIR fluorescence probes (MC-RdTPA1, MC-RdTPA2, and MC-RdTPE1) have been constructed. These probes have several advantages: (i) have long emission wavelength and large Stokes shifts, which have great potential in vivo imaging applications; (ii) could selectively visualize MC-LR in cells; and (iii) showed stable fluorescence intensity in the pH range of 5.0-7.0. This work may provide a new avenue for the detection of MC-LR in biological systems and new tool to advance our knowledge of the mechanism of MC-LR toxicity.
Collapse
Affiliation(s)
- Huiyan Chuan
- Institute for Ecological Research and Pollution Control of Plateau Lakes, School of Ecology and Environmental Sciences, Yunnan University, Kunming 650500, P. R. China
| | - Bingyan Li
- Institute for Ecological Research and Pollution Control of Plateau Lakes, School of Ecology and Environmental Sciences, Yunnan University, Kunming 650500, P. R. China
| | - Zhaomin Wang
- Institute for Ecological Research and Pollution Control of Plateau Lakes, School of Ecology and Environmental Sciences, Yunnan University, Kunming 650500, P. R. China
| | - Jing Li
- Yunnan International Joint R&D Center of Smart Agriculture and Water Security; School of Water Conservancy, Yunnan Agricultural University, Kunming 650201, P. R. China
| | - Ping Xie
- Institute for Ecological Research and Pollution Control of Plateau Lakes, School of Ecology and Environmental Sciences, Yunnan University, Kunming 650500, P. R. China
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, PR China
| | - Yong Liu
- Institute for Ecological Research and Pollution Control of Plateau Lakes, School of Ecology and Environmental Sciences, Yunnan University, Kunming 650500, P. R. China
| |
Collapse
|
26
|
Doha ZO, Sears RC. Unraveling MYC's Role in Orchestrating Tumor Intrinsic and Tumor Microenvironment Interactions Driving Tumorigenesis and Drug Resistance. PATHOPHYSIOLOGY 2023; 30:400-419. [PMID: 37755397 PMCID: PMC10537413 DOI: 10.3390/pathophysiology30030031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/28/2023] Open
Abstract
The transcription factor MYC plays a pivotal role in regulating various cellular processes and has been implicated in tumorigenesis across multiple cancer types. MYC has emerged as a master regulator governing tumor intrinsic and tumor microenvironment interactions, supporting tumor progression and driving drug resistance. This review paper aims to provide an overview and discussion of the intricate mechanisms through which MYC influences tumorigenesis and therapeutic resistance in cancer. We delve into the signaling pathways and molecular networks orchestrated by MYC in the context of tumor intrinsic characteristics, such as proliferation, replication stress and DNA repair. Furthermore, we explore the impact of MYC on the tumor microenvironment, including immune evasion, angiogenesis and cancer-associated fibroblast remodeling. Understanding MYC's multifaceted role in driving drug resistance and tumor progression is crucial for developing targeted therapies and combination treatments that may effectively combat this devastating disease. Through an analysis of the current literature, this review's goal is to shed light on the complexities of MYC-driven oncogenesis and its potential as a promising therapeutic target.
Collapse
Affiliation(s)
- Zinab O. Doha
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA;
- Department of Medical Laboratories Technology, Taibah University, Al-Madinah 42353, Saudi Arabia
| | - Rosalie C. Sears
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA;
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| |
Collapse
|
27
|
Peris I, Romero-Murillo S, Vicente C, Narla G, Odero MD. Regulation and role of the PP2A-B56 holoenzyme family in cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188953. [PMID: 37437699 DOI: 10.1016/j.bbcan.2023.188953] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/14/2023]
Abstract
Protein phosphatase 2A (PP2A) inactivation is common in cancer, leading to sustained activation of pro-survival and growth-promoting pathways. PP2A consists of a scaffolding A-subunit, a catalytic C-subunit, and a regulatory B-subunit. The functional complexity of PP2A holoenzymes arises mainly through the vast repertoire of regulatory B-subunits, which determine both their substrate specificity and their subcellular localization. Therefore, a major challenge for developing more effective therapeutic strategies for cancer is to identify the specific PP2A complexes to be targeted. Of note, the development of small molecules specifically directed at PP2A-B56α has opened new therapeutic avenues in both solid and hematological tumors. Here, we focus on the B56/PR61 family of PP2A regulatory subunits, which have a central role in directing PP2A tumor suppressor activity. We provide an overview of the mechanisms controlling the formation and regulation of these complexes, the pathways they control, and the mechanisms underlying their deregulation in cancer.
Collapse
Affiliation(s)
- Irene Peris
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain; Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| | - Silvia Romero-Murillo
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain; Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain
| | - Carmen Vicente
- Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, MI, USA
| | - Maria D Odero
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain; Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
28
|
Feng Y, Massarelli E, Forman E, Kovach JS, Salgia R, Synold TW. An LC-MS/MS method for simultaneous determination of LB-100 and its active metabolite, endothall, in human plasma. Bioanalysis 2023; 15:1095-1107. [PMID: 37584370 PMCID: PMC10505989 DOI: 10.4155/bio-2023-0078] [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: 05/01/2023] [Accepted: 07/13/2023] [Indexed: 08/17/2023] Open
Abstract
We have developed and validated a novel LC-MS/MS method for the simultaneous quantification of LB-100 and its active metabolite, endothall, in human plasma following solid-phase extraction. LB-105 and endothall-D6 were used as internal standards. Chromatographic separation was achieved on a Hypercarb™ column using 5 mM (NH4)2CO3 and 30:70 (v/v) 100 mM (NH4)2CO3:acetonitrile as mobile phases. Detection was performed via positive electrospray ionization mode with multiple reaction monitoring. The assay exhibited linearity in the concentration range of 2.5-500 ng/ml for both analytes. Intra- and inter-assay precision and accuracy were within ±11%. LB-100 and endothall recoveries were 78.7 and 86.7%, respectively. The validated LC-MS/MS method enabled the accurate measurement of LB-100 and endothall in patient samples from an ongoing clinical trial (NCT04560972).
Collapse
Affiliation(s)
- Ye Feng
- Department of Medical Oncology & Therapeutics Research, City of Hope, 1500 East Duarte Rd, Duarte, CA 91010, USA
| | - Erminia Massarelli
- Department of Medical Oncology & Therapeutics Research, City of Hope, 1500 East Duarte Rd, Duarte, CA 91010, USA
| | - Eric Forman
- LIXTE Biotechnology, Inc. 680 E Colorado Blvd, Suite 180, Pasadena, CA 91101, USA
| | - John S Kovach
- LIXTE Biotechnology, Inc. 680 E Colorado Blvd, Suite 180, Pasadena, CA 91101, USA
| | - Ravi Salgia
- Department of Medical Oncology & Therapeutics Research, City of Hope, 1500 East Duarte Rd, Duarte, CA 91010, USA
| | - Timothy W Synold
- Department of Medical Oncology & Therapeutics Research, City of Hope, 1500 East Duarte Rd, Duarte, CA 91010, USA
| |
Collapse
|
29
|
Domènech Omella J, Cortesi EE, Verbinnen I, Remmerie M, Wu H, Cubero FJ, Roskams T, Janssens V. A Novel Mouse Model of Combined Hepatocellular-Cholangiocarcinoma Induced by Diethylnitrosamine and Loss of Ppp2r5d. Cancers (Basel) 2023; 15:4193. [PMID: 37627221 PMCID: PMC10453342 DOI: 10.3390/cancers15164193] [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: 07/14/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Primary liver cancer (PLC) can be classified in hepatocellular (HCC), cholangiocarcinoma (CCA), and combined hepatocellular-cholangiocarcinoma (cHCC-CCA). The molecular mechanisms involved in PLC development and phenotype decision are still not well understood. Complete deletion of Ppp2r5d, encoding the B56δ subunit of Protein Phosphatase 2A (PP2A), results in spontaneous HCC development in mice via a c-MYC-dependent mechanism. In the present study, we aimed to examine the role of Ppp2r5d in an independent mouse model of diethylnitrosamine (DEN)-induced hepatocarcinogenesis. Ppp2r5d deletion (heterozygous and homozygous) accelerated HCC development, corroborating its tumor-suppressive function in liver and suggesting Ppp2r5d may be haploinsufficient. Ppp2r5d-deficient HCCs stained positively for c-MYC, consistent with increased AKT activation in pre-malignant and tumor tissues of Ppp2r5d-deficient mice. We also found increased YAP activation in Ppp2r5d-deficient tumors. Remarkably, in older mice, Ppp2r5d deletion resulted in cHCC-CCA development in this model, with the CCA component showing increased expression of progenitor markers (SOX9 and EpCAM). Finally, we observed an upregulation of Ppp2r5d in tumors from wildtype and heterozygous mice, revealing a tumor-specific control mechanism of Ppp2r5d expression, and suggestive of the involvement of Ppp2r5d in a negative feedback regulation restricting tumor growth. Our study highlights the tumor-suppressive role of mouse PP2A-B56δ in both HCC and cHCC-CCA, which may have important implications for human PLC development and targeted treatment.
Collapse
Affiliation(s)
- Judit Domènech Omella
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), 3000 Leuven, Belgium; (J.D.O.); (I.V.); (M.R.)
| | - Emanuela E. Cortesi
- Translational Cell & Tissue Research, University of Leuven (KU Leuven), 3000 Leuven, Belgium; (E.E.C.); (T.R.)
| | - Iris Verbinnen
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), 3000 Leuven, Belgium; (J.D.O.); (I.V.); (M.R.)
| | - Michiel Remmerie
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), 3000 Leuven, Belgium; (J.D.O.); (I.V.); (M.R.)
| | - Hanghang Wu
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, 28040 Madrid, Spain; (H.W.); (F.J.C.)
| | - Francisco J. Cubero
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, 28040 Madrid, Spain; (H.W.); (F.J.C.)
- Health Research Institute Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Centre for Biomedical Research, Network on Liver and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain
| | - Tania Roskams
- Translational Cell & Tissue Research, University of Leuven (KU Leuven), 3000 Leuven, Belgium; (E.E.C.); (T.R.)
- Department of Pathology, University Hospitals Leuven (UZ Leuven), 3000 Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), 3000 Leuven, Belgium; (J.D.O.); (I.V.); (M.R.)
- KU Leuven Cancer Institute (LKI), 3000 Leuven, Belgium
| |
Collapse
|
30
|
Kim H, Takegahara N, Choi Y. PP2A-Mediated GSK3β Dephosphorylation Is Required for Protocadherin-7-Dependent Regulation of Small GTPase RhoA in Osteoclasts. Cells 2023; 12:1967. [PMID: 37566044 PMCID: PMC10417323 DOI: 10.3390/cells12151967] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023] Open
Abstract
Protocadherin-7 (Pcdh7) is a member of the non-clustered protocadherin δ1 subgroup of the cadherin superfamily. Pcdh7 has been revealed to control osteoclast differentiation by regulating Rho-family small GTPases, RhoA and Rac1, through its intracellular SET binding domain. However, the mechanisms by which small GTPases are regulated downstream of Pcdh7 remain unclear. Here, we demonstrate that protein phosphatase 2A (PP2A)-mediated dephosphorylation of Glycogen synthase kinase-3β (GSK3β) is required for Pcdh7-dependent activation of RhoA during osteoclast differentiation. Pcdh7-deficient (Pcdh7-/-) cells showed impaired PP2A activity, despite their normal expression of PP2A. GSK3β, whose activity is regulated by its inhibitory phosphorylation at Ser9, was dephosphorylated during osteoclast differentiation in a Pcdh7-dependent manner. Inhibition of protein phosphatase by okadaic acid reduced dephosphorylation of GSK3β in Pcdh7+/+ cells, while activation of PP2A by DT-061 rescued impaired dephosphorylation of GSK3β in Pcdh7-/- cells. Inhibition of GSK3β by AR-A014418 inhibited RANKL-induced RhoA activation and osteoclast differentiation in Pcdh7+/+ cells. On the other hand, DT-061 treatment rescued impaired RhoA activation and RANKL-induced osteoclast differentiation in Pcdh7-/- cells. Taken together, these results demonstrate that PP2A dephosphorylates GSK3β and thereby activates it in a Pcdh7-dependent manner, which is required for activation of small GTPase RhoA and proper osteoclast differentiation.
Collapse
Affiliation(s)
| | | | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; (H.K.); (N.T.)
| |
Collapse
|
31
|
Guffens L, Derua R, Janssens V. PME-1 sensitizes glioblastoma cells to oxidative stress-induced cell death by attenuating PP2A-B55α-mediated inactivation of MAPKAPK2-RIPK1 signaling. Cell Death Discov 2023; 9:265. [PMID: 37500619 PMCID: PMC10374899 DOI: 10.1038/s41420-023-01572-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/13/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023] Open
Abstract
Glioblastoma (GBM) is the most common primary brain tumor in adults. Current standard therapy is surgery followed by radiotherapy, with concurrent and adjuvant temozolomide chemotherapy. GBM is characterized by almost uniformly fatal outcomes, highlighting the unmet clinical need for more efficient, biomarker-guided treatments. Protein phosphatase methylesterase-1 (PME-1), a regulator of the tumor suppressive phosphatase PP2A, promotes PP2A demethylation and inactivation, and is overexpressed in 44% of GBM, associated with increased tumor grade and cellular proliferation. Here, we aimed to investigate how reactive oxygen species (ROS), a frequent by-product of radiotherapy and temozolomide chemotherapy, regulate PP2A function via its methylesterase PME-1, and how PME-1 overexpression impacts the response of GBM cells to oxidative stress. We found that in two glioblastoma cell lines, U87MG and U251MG, expression of PME-1 is positively correlated with the sensitivity of the cells to H2O2 or t-BHP-induced oxidative stress. Experiments using the irreversible pharmacologic PME-1 inhibitor, AMZ30, and different PME-1 mutants, revealed that the methylesterase function, the PP2A binding capacity, and the nuclear localization of PME-1 are all important for the sensitizing effect of PME-1 expression. Furthermore, we identified increased nuclear localization of the PP2A-B55α subunit, increased binding of PP2A-B55α to PME-1, and increased B55α-bound PP2A-C demethylation upon oxidative stress. Lastly, we uncovered increased stress-induced phosphorylation and activity of MAPKAPK2 and RIPK1 in PME-1 overexpressing U87MG cells, which caused the observed sensitization to t-BHP treatment. Our data reveal a novel role for PME-1 in oxidative stress-induced GBM cell death, regulating nuclear PP2A-B55α activity and MAPKAPK2-RIPK1 signaling. Patients with GBM tumors overexpressing PME-1, although having a worse prognosis due to increased cellular proliferation of the tumor, could actually be more responsive to oxidative stress-inducing therapies.
Collapse
Affiliation(s)
- Liesbeth Guffens
- Laboratory of Protein Phosphorylation & Proteomics, Dept. Cellular & Molecular Medicine, KU Leuven, B-3000, Leuven, Belgium
- KU Leuven Cancer Institute (LKI), B-3000, Leuven, Belgium
| | - Rita Derua
- Laboratory of Protein Phosphorylation & Proteomics, Dept. Cellular & Molecular Medicine, KU Leuven, B-3000, Leuven, Belgium
- SyBioMa, KU Leuven, B-3000, Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation & Proteomics, Dept. Cellular & Molecular Medicine, KU Leuven, B-3000, Leuven, Belgium.
- KU Leuven Cancer Institute (LKI), B-3000, Leuven, Belgium.
| |
Collapse
|
32
|
Liu H, Fang D, Zhang C, Zhao Z, Liu Y, Zhao S, Zhang N, Xu J. Circular MTHFD2L RNA-encoded CM-248aa inhibits gastric cancer progression by targeting the SET-PP2A interaction. Mol Ther 2023; 31:1739-1755. [PMID: 37101395 PMCID: PMC10277894 DOI: 10.1016/j.ymthe.2023.04.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 01/10/2023] [Accepted: 04/21/2023] [Indexed: 04/28/2023] Open
Abstract
The available targeted therapies for gastric cancer (GC) are still limited, so it is important to discover novel molecules as potential treatment options. Proteins or peptides encoded by circular RNAs (circRNAs) are increasingly reported to play essential roles in malignancies. The aim of the present study was to identify an undiscovered protein encoded by circRNA and explore its key role and molecular mechanism in GC progression. CircMTHFD2L (hsa_circ_0069982) was screened and validated as a downregulated circRNA with coding potential. The protein encoded by circMTHFD2L, named CM-248aa, was identified for the first time by immunoprecipitation and mass spectrometry. CM-248aa was significantly downregulated in GC, while its low expression was associated with advanced tumor-node-metastasis (TNM) stage and histopathological grade. Low expression of CM-248aa could be an independent risk factor for poor prognosis. Functionally, CM-248aa, instead of circMTHFD2L suppressed the proliferation and metastasis of GC in vitro and in vivo. Mechanistically, CM-248aa competitively targeted the acidic domain of SET nuclear oncogene (SET) and acted as an endogenous inhibitor of the SET-protein phosphatase 2A interaction to promote dephosphorylation of AKT, extracellular signal-regulated kinase, and P65. Our discovery revealed that CM-248aa could be a potential prognostic biomarker and endogenous therapeutic option for GC.
Collapse
Affiliation(s)
- Haohan Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan 2 Road, Guangzhou, Guangdong 510080, People's Republic of China; Laboratory of General Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan 2 Road, Guangzhou, Guangdong 510080, People's Republic of China
| | - Deliang Fang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan 2 Road, Guangzhou, Guangdong 510080, People's Republic of China; Laboratory of General Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan 2 Road, Guangzhou, Guangdong 510080, People's Republic of China
| | - Chaoyue Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan 2 Road, Guangzhou, Guangdong 510080, People's Republic of China
| | - Zirui Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan 2 Road, Guangzhou, Guangdong 510080, People's Republic of China
| | - Yinan Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan 2 Road, Guangzhou, Guangdong 510080, People's Republic of China; Laboratory of General Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan 2 Road, Guangzhou, Guangdong 510080, People's Republic of China
| | - Shaoji Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan 2 Road, Guangzhou, Guangdong 510080, People's Republic of China
| | - Nu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan 2 Road, Guangzhou, Guangdong 510080, People's Republic of China.
| | - Jianbo Xu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan 2 Road, Guangzhou, Guangdong 510080, People's Republic of China.
| |
Collapse
|
33
|
Zhong Q, Xiao X, Qiu Y, Xu Z, Chen C, Chong B, Zhao X, Hai S, Li S, An Z, Dai L. Protein posttranslational modifications in health and diseases: Functions, regulatory mechanisms, and therapeutic implications. MedComm (Beijing) 2023; 4:e261. [PMID: 37143582 PMCID: PMC10152985 DOI: 10.1002/mco2.261] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
Protein posttranslational modifications (PTMs) refer to the breaking or generation of covalent bonds on the backbones or amino acid side chains of proteins and expand the diversity of proteins, which provides the basis for the emergence of organismal complexity. To date, more than 650 types of protein modifications, such as the most well-known phosphorylation, ubiquitination, glycosylation, methylation, SUMOylation, short-chain and long-chain acylation modifications, redox modifications, and irreversible modifications, have been described, and the inventory is still increasing. By changing the protein conformation, localization, activity, stability, charges, and interactions with other biomolecules, PTMs ultimately alter the phenotypes and biological processes of cells. The homeostasis of protein modifications is important to human health. Abnormal PTMs may cause changes in protein properties and loss of protein functions, which are closely related to the occurrence and development of various diseases. In this review, we systematically introduce the characteristics, regulatory mechanisms, and functions of various PTMs in health and diseases. In addition, the therapeutic prospects in various diseases by targeting PTMs and associated regulatory enzymes are also summarized. This work will deepen the understanding of protein modifications in health and diseases and promote the discovery of diagnostic and prognostic markers and drug targets for diseases.
Collapse
Affiliation(s)
- Qian Zhong
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Xina Xiao
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Yijie Qiu
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Zhiqiang Xu
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Chunyu Chen
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Baochen Chong
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Xinjun Zhao
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Shan Hai
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Shuangqing Li
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Zhenmei An
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Lunzhi Dai
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
34
|
Roy S, Batra L. Protein Phosphatase 2A: Role in T Cells and Diseases. J Immunol Res 2023; 2023:4522053. [PMID: 37234102 PMCID: PMC10208765 DOI: 10.1155/2023/4522053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/27/2023] Open
Abstract
Protein phosphatase 2A (PP2A) is a serine-threonine phosphatase that plays an important role in the regulation of cell proliferation and signal transduction. The catalytic activity of PP2A is integral in the maintenance of physiological functions which gets severely impaired in its absence. PP2A plays an essential role in the activation, differentiation, and functions of T cells. PP2A suppresses Th1 cell differentiation while promoting Th2 cell differentiation. PP2A fosters Th17 cell differentiation which contributes to the pathogenesis of systemic lupus erythematosus (SLE) by enhancing the transactivation of the Il17 gene. Genetic deletion of PP2A in Tregs disrupts Foxp3 expression due to hyperactivation of mTORC1 signaling which impairs the development and immunosuppressive functions of Tregs. PP2A is important in the induction of Th9 cells and promotes their antitumor functions. PP2A activation has shown to reduce neuroinflammation in a mouse model of experimental autoimmune encephalomyelitis (EAE) and is now used to treat multiple sclerosis (MS) clinically. In this review, we will discuss the structure and functions of PP2A in T cell differentiation and diseases and therapeutic applications of PP2A-mediated immunotherapy.
Collapse
Affiliation(s)
- Suyasha Roy
- Immuno-Biology Laboratory, Translational Health Science and Technology Institute, Faridabad, India
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lalit Batra
- Regional Biocontainment Laboratory, Center for Predictive Medicine, University of Louisville, Louisville, KY, USA
| |
Collapse
|
35
|
Zhao W, Xu C, Peng L, Ma L, Du M. cAMP/PKA signaling promotes AKT deactivation by reducing CIP2A expression, thereby facilitating decidualization. Mol Cell Endocrinol 2023; 571:111946. [PMID: 37127088 DOI: 10.1016/j.mce.2023.111946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/24/2023] [Accepted: 04/28/2023] [Indexed: 05/03/2023]
Abstract
cAMP signaling is widely known to be indispensable for decidualization, but the details are not fully understood. Here, we show that cAMP signaling promotes AKT deactivation in endometrial stromal cells, which favors their decidualization. The deactivation of AKT is found to be a consequence of the reduced expression of several inhibitors of PP2A, the major phosphatase of AKT, with CIP2A being the most prominent. CIP2A reduction is obligatory for decidualization, as persistent CIP2A expression impairs chromatin remodeling and the expression of several decidualization markers (IGFBP1, PRL, MAOA, and IL-15). Furthermore, analyses of the responsiveness of the CIP2A promoter to cAMP signaling suggest the ETS family to be a bridge between cAMP signaling and CIP2A reduction. Our results provide novel insights into the role of cAMP signaling in decidualization and might benefit the development of novel therapies for decidualization deficiency, AKT-driven tumors, and the reverse, insulin resistance.
Collapse
Affiliation(s)
- Weijie Zhao
- Reproductive Medicine Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Chunfang Xu
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, Shanghai, 200090, China
| | - Lijin Peng
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, Shanghai, 200090, China
| | - Lin Ma
- Reproductive Medicine Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
| | - Meirong Du
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, Shanghai, 200090, China.
| |
Collapse
|
36
|
Marin GH, Murail S, Andrini L, Garcia M, Loisel S, Tuffery P, Rebollo A. In Silico and In Vivo Studies of a Tumor-Penetrating and Interfering Peptide with Antitumoral Effect on Xenograft Models of Breast Cancer. Pharmaceutics 2023; 15:pharmaceutics15041180. [PMID: 37111665 PMCID: PMC10142558 DOI: 10.3390/pharmaceutics15041180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/09/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
The combination of a tumor-penetrating peptide (TPP) with a peptide able to interfere with a given protein-protein interaction (IP) is a promising strategy with potential clinical application. Little is known about the impact of fusing a TPP with an IP, both in terms of internalization and functional effect. Here, we analyze these aspects in the context of breast cancer, targeting PP2A/SET interaction, using both in silico and in vivo approaches. Our results support the fact that state-of-the-art deep learning approaches developed for protein-peptide interaction modeling can reliably identify good candidate poses for the IP-TPP in interaction with the Neuropilin-1 receptor. The association of the IP with the TPP does not seem to affect the ability of the TPP to bind to Neuropilin-1. Molecular simulation results suggest that peptide IP-GG-LinTT1 in a cleaved form interacts with Neuropilin-1 in a more stable manner and has a more helical secondary structure than the cleaved IP-GG-iRGD. Surprisingly, in silico investigations also suggest that the non-cleaved TPPs can bind the Neuropilin-1 in a stable manner. The in vivo results using xenografts models show that both bifunctional peptides resulting from the combination of the IP and either LinTT1 or iRGD are effective against tumoral growth. The peptide iRGD-IP shows the highest stability to serum proteases degradation while having the same antitumoral effect as Lin TT1-IP, which is more sensitive to proteases degradation. Our results support the development of the TPP-IP strategy as therapeutic peptides against cancer.
Collapse
Affiliation(s)
- Gustavo H Marin
- Department of Pharmacology/Histology and Embryology, FMC, National University of La Plata, CONICET, La Plata 1900, Argentina
| | - Samuel Murail
- BFA, Université Paris Cite, CNRS UMR 8251, Inserm U1133, 75013 Paris, France
| | - Laura Andrini
- Department of Pharmacology/Histology and Embryology, FMC, National University of La Plata, CONICET, La Plata 1900, Argentina
| | - Marcela Garcia
- Department of Pharmacology/Histology and Embryology, FMC, National University of La Plata, CONICET, La Plata 1900, Argentina
| | | | - Pierre Tuffery
- BFA, Université Paris Cite, CNRS UMR 8251, Inserm U1133, 75013 Paris, France
| | - Angelita Rebollo
- Faculté de Pharmacie, UTCBS, Université Paris Cite, Inserm U1267, 75006 Paris, France
| |
Collapse
|
37
|
Wilde L, Porazzi P, Trotta R, De Dominici M, Palmisiano N, Keiffer G, Rancani K, Yingling K, Calabretta B, Kasner M. A phase I study of the combination of palbociclib and dexamethasone for the treatment of relapsed or refractory B-cell acute lymphoblastic leukemia. Leuk Res 2023; 129:107075. [PMID: 37079999 DOI: 10.1016/j.leukres.2023.107075] [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: 12/21/2022] [Revised: 03/20/2023] [Accepted: 04/05/2023] [Indexed: 04/22/2023]
Abstract
PURPOSE Despite advances in the treatment of B-cell acute lymphoblastic leukemia (B-ALL), outcomes for relapsed/refractory (R/R) disease remain poor. Preclinical studies suggest that the combination of the CDK4/6 inhibitor palbociclib and dexamethasone may be effective in targeting leukemic cell growth. We conducted a phase I study of escalating doses of palbociclib in combination with dexamethasone in adults with R/R B-ALL. METHODS Cycle 1 consisted of single agent palbociclib given for 7 days and continued for 28 additional days in combination with dexamethasone 20 mg daily. Palbociclib dosing began at 100 mg daily. Patients with a response were eligible for maintenance consisting of 1 week of palbociclib plus dexamethasone (20 mg daily × 2 days, 16 mg daily × 2 days, 12 mg daily × 2 days, 6 mg daily × 1 day), followed by 3 weeks of palbociclib alone. Safety, efficacy, and the expression of phospho-RB and c-MYB/BCL-2 were measured. CONCLUSIONS Seven patients were treated on study before it was closed early due to slow accrual. No dose limiting toxicities were identified. One patient had a complete response with incomplete hematologic recovery, suggesting possible efficacy of the treatment. Reduction in CD34+ cells, p-RB, c-MYB, and BCL-2 expression also suggested on-target therapy effects.
Collapse
Affiliation(s)
- Lindsay Wilde
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, United States.
| | - Patrizia Porazzi
- Division of Hematology and Oncology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, United States
| | - Rossana Trotta
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Marco De Dominici
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, CO, United States
| | - Neil Palmisiano
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, United States
| | - Gina Keiffer
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, United States
| | - Kaitlin Rancani
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, United States
| | - Kathryn Yingling
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, United States
| | - Bruno Calabretta
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Margaret Kasner
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, United States
| |
Collapse
|
38
|
Kim J, Ji S, Lee JY, Lorquin J, Orlikova-Boyer B, Cerella C, Mazumder A, Muller F, Dicato M, Detournay O, Diederich M. Marine Polyether Phycotoxin Palytoxin Induces Apoptotic Cell Death via Mcl-1 and Bcl-2 Downregulation. Mar Drugs 2023; 21:md21040233. [PMID: 37103372 PMCID: PMC10143546 DOI: 10.3390/md21040233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 04/28/2023] Open
Abstract
Palytoxin is considered one of the most potent biotoxins. As palytoxin-induced cancer cell death mechanisms remain to be elucidated, we investigated this effect on various leukemia and solid tumor cell lines at low picomolar concentrations. As palytoxin did not affect the viability of peripheral blood mononuclear cells (PBMC) from healthy donors and did not create systemic toxicity in zebrafish, we confirmed excellent differential toxicity. Cell death was characterized by a multi-parametric approach involving the detection of nuclear condensation and caspase activation assays. zVAD-sensitive apoptotic cell death was concomitant with a dose-dependent downregulation of antiapoptotic Bcl-2 family proteins Mcl-1 and Bcl-xL. Proteasome inhibitor MG-132 prevented the proteolysis of Mcl-1, whereas the three major proteasomal enzymatic activities were upregulated by palytoxin. Palytoxin-induced dephosphorylation of Bcl-2 further exacerbated the proapoptotic effect of Mcl-1 and Bcl-xL degradation in a range of leukemia cell lines. As okadaic acid rescued cell death triggered by palytoxin, protein phosphatase (PP)2A was involved in Bcl-2 dephosphorylation and induction of apoptosis by palytoxin. At a translational level, palytoxin abrogated the colony formation capacity of leukemia cell types. Moreover, palytoxin abrogated tumor formation in a zebrafish xenograft assay at concentrations between 10 and 30 pM. Altogether, we provide evidence of the role of palytoxin as a very potent and promising anti-leukemic agent, acting at low picomolar concentrations in cellulo and in vivo.
Collapse
Affiliation(s)
- Jaemyun Kim
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Republic of Korea
| | - Seungwon Ji
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Republic of Korea
| | - Jin-Young Lee
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Republic of Korea
| | - Jean Lorquin
- Institut Méditerranéen d'Océanologie, 163 Avenue de Luminy, CEDEX 09, 13288 Marseille, France
| | - Barbora Orlikova-Boyer
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Republic of Korea
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer (LBMCC), BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210 Luxembourg, Luxembourg
| | - Claudia Cerella
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Republic of Korea
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer (LBMCC), BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210 Luxembourg, Luxembourg
| | - Aloran Mazumder
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Republic of Korea
| | - Florian Muller
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer (LBMCC), BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210 Luxembourg, Luxembourg
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer (LBMCC), BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210 Luxembourg, Luxembourg
| | - Olivier Detournay
- Planktovie SAS, 45 Rue Frédéric Joliot Curie, CEDEX 13, 13013 Marseille, France
| | - Marc Diederich
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Republic of Korea
| |
Collapse
|
39
|
Stanford SM, Bottini N. Targeting protein phosphatases in cancer immunotherapy and autoimmune disorders. Nat Rev Drug Discov 2023; 22:273-294. [PMID: 36693907 PMCID: PMC9872771 DOI: 10.1038/s41573-022-00618-w] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2022] [Indexed: 01/25/2023]
Abstract
Protein phosphatases act as key regulators of multiple important cellular processes and are attractive therapeutic targets for various diseases. Although extensive effort has been dedicated to phosphatase-targeted drug discovery, early expeditions for competitive phosphatase inhibitors were plagued by druggability issues, leading to the stigmatization of phosphatases as difficult targets. Despite challenges, persistent efforts have led to the identification of several drug-like, non-competitive modulators of some of these enzymes - including SH2 domain-containing protein tyrosine phosphatase 2, protein tyrosine phosphatase 1B, vascular endothelial protein tyrosine phosphatase and protein phosphatase 1 - reigniting interest in therapeutic targeting of phosphatases. Here, we discuss recent progress in phosphatase drug discovery, with emphasis on the development of selective modulators that exhibit biological activity. The roles and regulation of protein phosphatases in immune cells and their potential as powerful targets for immuno-oncology and autoimmunity indications are assessed.
Collapse
Affiliation(s)
| | - Nunzio Bottini
- Department of Medicine, University of California, San Diego, CA, USA.
| |
Collapse
|
40
|
Ho WS, Mondal I, Xu B, Das O, Sun R, Chiou P, Cai X, Tahmasebinia F, McFadden E, Wu CYJ, Wu Z, Matsui W, Lim M, Meng Z, Lu RO. PP2Ac/STRN4 negatively regulates STING-type I IFN signaling in tumor-associated macrophages. J Clin Invest 2023; 133:e162139. [PMID: 36757811 PMCID: PMC10014107 DOI: 10.1172/jci162139] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
Stimulator of IFN genes type I (STING-Type I) IFN signaling in myeloid cells plays a critical role in effective antitumor immune responses, but STING agonists as monotherapy have shown limited efficacy in clinical trials. The mechanisms that downregulate STING signaling are not fully understood. Here, we report that protein phosphatase 2A (PP2A), with its specific B regulatory subunit Striatin 4 (STRN4), negatively regulated STING-Type I IFN in macrophages. Mice with macrophage PP2A deficiency exhibited reduced tumor progression. The tumor microenvironment showed decreased immunosuppressive and increased IFN-activated macrophages and CD8+ T cells. Mechanistically, we demonstrated that Hippo kinase MST1/2 was required for STING activation. STING agonists induced dissociation of PP2A from MST1/2 in normal macrophages, but not in tumor conditioned macrophages. Furthermore, our data showed that STRN4 mediated PP2A binding to and dephosphorylation of Hippo kinase MST1/2, resulting in stabilization of YAP/TAZ to antagonize STING activation. In human patients with glioblastoma (GBM), YAP/TAZ was highly expressed in tumor-associated macrophages but not in nontumor macrophages. We also demonstrated that PP2A/STRN4 deficiency in macrophages reduced YAP/TAZ expression and sensitized tumor-conditioned macrophages to STING stimulation. In summary, we demonstrated that PP2A/STRN4-YAP/TAZ has, in our opinion, been an unappreciated mechanism that mediates immunosuppression in tumor-associated macrophages, and targeting the PP2A/STRN4-YAP/TAZ axis can sensitize tumors to immunotherapy.
Collapse
Affiliation(s)
- Winson S. Ho
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Isha Mondal
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Beisi Xu
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Oishika Das
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Raymond Sun
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Pochin Chiou
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Xiaomin Cai
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Foozhan Tahmasebinia
- Department of Biological Sciences, Southern Methodist University, Dallas, Texas, USA
| | - Elizabeth McFadden
- Department of Molecular Sciences, University of Texas at Austin, Austin, Texas, USA
| | - Caren Yu-Ju Wu
- Department of Neurosurgery, Stanford University, Stanford, California, USA
| | - Zhihao Wu
- Department of Biological Sciences, Southern Methodist University, Dallas, Texas, USA
| | - William Matsui
- Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Michael Lim
- Department of Neurosurgery, Stanford University, Stanford, California, USA
| | - Zhipeng Meng
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Rongze Olivia Lu
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
- Helen Diller Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| |
Collapse
|
41
|
Zhou Y, Fan K, Dou N, Li L, Wang J, Chen J, Li Y, Gao Y. YTHDF2 exerts tumor-suppressor roles in gastric cancer via up-regulating PPP2CA independently of m 6A modification. Biol Proced Online 2023; 25:6. [PMID: 36870954 PMCID: PMC9985201 DOI: 10.1186/s12575-023-00195-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND YTHDF2 is one of important readers of N6-methyladenosine (m6A) modification on RNA. Growing evidence implicates that YTHDF2 takes an indispensable part in the regulation of tumorigenesis and metastasis in different cancers, but its biological functions and underlying mechanisms remain elusive in gastric cancer (GC). AIM To investigate the clinical relevance and biological function of YTHDF2 in GC. RESULTS Compared with matched normal stomach tissues, YTHDF2 expression was markedly decreased in gastric cancer tissues. The expression level of YTHDF2 was inversely associated with gastric cancer patients' tumor size, AJCC classification and prognosis. Functionally, YTHDF2 reduction facilitated gastric cancer cell growth and migration in vitro and in vivo, whereas YTHDF2 overexpression exhibited opposite phenotypes. Mechanistically, YTHDF2 enhanced expression of PPP2CA, the catalytic subunit of PP2A (Protein phosphatase 2A), in an m6A-independent manner, and silencing of PPP2CA antagonized the anti-tumor effects caused by overexpression of YTHDF2 in GC cells. CONCLUSION These findings demonstrate that YTHDF2 is down-regulated in GC and its down-regulation promotes GC progression via a possible mechanism involving PPP2CA expression, suggesting that YTHDF2 may be a hopeful biomarker for diagnosis and an unrevealed treatment target for GC.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, 150 Ji-Mo Rd., Shanghai, 200120, China
| | - Kailing Fan
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, 150 Ji-Mo Rd., Shanghai, 200120, China
| | - Ning Dou
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, 150 Ji-Mo Rd., Shanghai, 200120, China
| | - Li Li
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, 150 Ji-Mo Rd., Shanghai, 200120, China
| | - Jialin Wang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, 150 Ji-Mo Rd., Shanghai, 200120, China
| | - Jingde Chen
- Department of Oncology, Shanghai East Hospital Ji'an Hospital, Ji'an City, 343000, Jiangxi Province, China.,School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yandong Li
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, 150 Ji-Mo Rd., Shanghai, 200120, China.
| | - Yong Gao
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, 150 Ji-Mo Rd., Shanghai, 200120, China.
| |
Collapse
|
42
|
Peris I, Romero-Murillo S, Martínez-Balsalobre E, Farrington CC, Arriazu E, Marcotegui N, Jiménez-Muñoz M, Alburquerque-Prieto C, Torres-López A, Fresquet V, Martínez-Climent JA, Mateos MC, Cayuela ML, Narla G, Odero MD, Vicente C. Activation of the PP2A-B56α heterocomplex synergizes with venetoclax therapies in AML through BCL2 and MCL1 modulation. Blood 2023; 141:1047-1059. [PMID: 36455198 PMCID: PMC10023731 DOI: 10.1182/blood.2022016466] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 11/18/2022] [Accepted: 11/18/2022] [Indexed: 12/02/2022] Open
Abstract
Venetoclax combination therapies are becoming the standard of care in acute myeloid leukemia (AML). However, the therapeutic benefit of these drugs in older/unfit patients is limited to only a few months, highlighting the need for more effective therapies. Protein phosphatase 2A (PP2A) is a tumor suppressor phosphatase with pleiotropic functions that becomes inactivated in ∼70% of AML cases. PP2A promotes cancer cell death by modulating the phosphorylation state in a variety of proteins along the mitochondrial apoptotic pathway. We therefore hypothesized that pharmacological PP2A reactivation could increase BCL2 dependency in AML cells and, thus, potentiate venetoclax-induced cell death. Here, by using 3 structurally distinct PP2A-activating drugs, we show that PP2A reactivation synergistically enhances venetoclax activity in AML cell lines, primary cells, and xenograft models. Through the use of gene editing tools and pharmacological approaches, we demonstrate that the observed therapeutic synergy relies on PP2A complexes containing the B56α regulatory subunit, of which expression dictates response to the combination therapy. Mechanistically, PP2A reactivation enhances venetoclax-driven apoptosis through simultaneous inhibition of antiapoptotic BCL2 and extracellular signal-regulated kinase signaling, with the latter decreasing MCL1 protein stability. Finally, PP2A targeting increases the efficacy of the clinically approved venetoclax and azacitidine combination in vitro, in primary cells, and in an AML patient-derived xenograft model. These preclinical results provide a scientific rationale for testing PP2A-activating drugs with venetoclax combinations in AML.
Collapse
Affiliation(s)
- Irene Peris
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Silvia Romero-Murillo
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain
| | - Elena Martínez-Balsalobre
- Cancer and Aging Group, Hospital Universitario Virgen de la Arrixaca, and Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
| | - Caroline C. Farrington
- Division of Genetic Medicine, Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, MI
| | - Elena Arriazu
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Oncología, Instituto de Salud Carlos III, Madrid, Spain
| | - Nerea Marcotegui
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
| | - Marta Jiménez-Muñoz
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
| | | | | | - Vicente Fresquet
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Oncología, Instituto de Salud Carlos III, Madrid, Spain
| | - Jose A. Martínez-Climent
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Oncología, Instituto de Salud Carlos III, Madrid, Spain
| | - Maria C. Mateos
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Hematology Service, Hospital Universitario de Navarra, Pamplona, Spain
| | - Maria L. Cayuela
- Cancer and Aging Group, Hospital Universitario Virgen de la Arrixaca, and Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, MI
| | - Maria D. Odero
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Oncología, Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Vicente
- Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| |
Collapse
|
43
|
Welsh SA, Gardini A. Genomic regulation of transcription and RNA processing by the multitasking Integrator complex. Nat Rev Mol Cell Biol 2023; 24:204-220. [PMID: 36180603 PMCID: PMC9974566 DOI: 10.1038/s41580-022-00534-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2022] [Indexed: 11/09/2022]
Abstract
In higher eukaryotes, fine-tuned activation of protein-coding genes and many non-coding RNAs pivots around the regulated activity of RNA polymerase II (Pol II). The Integrator complex is the only Pol II-associated large multiprotein complex that is metazoan specific, and has therefore been understudied for years. Integrator comprises at least 14 subunits, which are grouped into distinct functional modules. The phosphodiesterase activity of the core catalytic module is co-transcriptionally directed against several RNA species, including long non-coding RNAs (lncRNAs), U small nuclear RNAs (U snRNAs), PIWI-interacting RNAs (piRNAs), enhancer RNAs and nascent pre-mRNAs. Processing of non-coding RNAs by Integrator is essential for their biogenesis, and at protein-coding genes, Integrator is a key modulator of Pol II promoter-proximal pausing and transcript elongation. Recent studies have identified an Integrator-specific serine/threonine-protein phosphatase 2A (PP2A) module, which targets Pol II and other components of the basal transcription machinery. In this Review, we discuss how the activity of Integrator regulates transcription, RNA processing, chromatin landscape and DNA repair. We also discuss the diverse roles of Integrator in development and tumorigenesis.
Collapse
|
44
|
Zhou X, Li C, Chen T, Li W, Wang X, Yang Q. Targeting RNA N6-methyladenosine to synergize with immune checkpoint therapy. Mol Cancer 2023; 22:36. [PMID: 36810108 PMCID: PMC9942356 DOI: 10.1186/s12943-023-01746-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/11/2023] [Indexed: 02/24/2023] Open
Abstract
Cancer immunotherapy, especially immune checkpoint therapy, has revolutionized therapeutic options by reactivating the host immune system. However, the efficacy varies, and only a small portion of patients develop sustained antitumor responses. Hence, illustrating novel strategies that improve the clinical outcome of immune checkpoint therapy is urgently needed. N6-methyladenosine (m6A) has been proved to be an efficient and dynamic posttranscriptional modification process. It is involved in numerous RNA processing, such as splicing, trafficking, translation and degradation. Compelling evidence emphasizes the paramount role of m6A modification in the regulation of immune response. These findings may provide a foundation for the rational combination of targeting m6A modification and immune checkpoints in cancer treatment. In the present review, we summarize the current landscape of m6A modification in RNA biology, and highlight the latest findings on the complex mechanisms by which m6A modification governs immune checkpoint molecules. Furthermore, given the critical role of m6A modification in antitumor immunity, we discuss the clinical significance of targeting m6A modification to improve the efficacy of immune checkpoint therapy for cancer control.
Collapse
Affiliation(s)
- Xianyong Zhou
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Wenhua Xi Road No. 107, Jinan, Shandong China ,grid.476866.dDepartment of Breast Surgery, Binzhou People’s Hospital, Binzhou, Shandong China
| | - Chen Li
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Wenhua Xi Road No. 107, Jinan, Shandong China
| | - Tong Chen
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Wenhua Xi Road No. 107, Jinan, Shandong China
| | - Wenhao Li
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Wenhua Xi Road No. 107, Jinan, Shandong China
| | - Xiaolong Wang
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Wenhua Xi Road No. 107, Jinan, Shandong, China.
| | - Qifeng Yang
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Wenhua Xi Road No. 107, Jinan, Shandong, China. .,Department of Pathology Tissue Bank, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Xi Road No. 107, Shandong, Jinan, China. .,Research Institute of Breast Cancer, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
45
|
Senapati J, Jabbour E, Kantarjian H, Short NJ. Pathogenesis and management of accelerated and blast phases of chronic myeloid leukemia. Leukemia 2023; 37:5-17. [PMID: 36309558 DOI: 10.1038/s41375-022-01736-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/11/2022] [Accepted: 10/14/2022] [Indexed: 02/01/2023]
Abstract
The treatment of chronic myeloid leukemia (CML) with tyrosine kinase inhibitors (TKIs) has been a model for cancer therapy development. Though most patients with CML have a normal quality and duration of life with TKI therapy, some patients progress to accelerated phase (AP) and blast phase (BP), both of which have a relatively poor prognosis. The rates of progression have reduced significantly from over >20% in the pre-TKI era to <5% now, largely due to refinements in CML therapy and response monitoring. Significant insights have been gained into the mechanisms of disease transformation including the role of additional cytogenetic abnormalities, somatic mutations, and other genomic alterations present at diagnosis or evolving on therapy. This knowledge is helping to optimize TKI therapy, improve prognostication and inform the development of novel combination regimens in these patients. While patients with de novo CML-AP have outcomes almost similar to CML in chronic phase (CP), those transformed from previously treated CML-CP should receive second- or third- generation TKIs and be strongly considered for allogeneic stem cell transplantation (allo-SCT). Similarly, patients with transformed CML-BP have particularly dismal outcomes with a median survival usually less than one year. Combination regimens with a potent TKI such as ponatinib followed by allo-SCT can achieve long-term survival in some transformed BP patients. Regimens including venetoclax in myeloid BP or inotuzumab ozogamicin or blinatumomab in lymphoid BP might lead to deeper and longer responses, facilitating potentially curative allo-SCT for patients with CML-BP once CP is achieved. Newer agents and novel combination therapies are further expanding the therapeutic arsenal in advanced phase CML.
Collapse
Affiliation(s)
- Jayastu Senapati
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
46
|
Chen B, Hu H, Chen X. From Basic Science to Clinical Practice: The Role of Cancerous Inhibitor of Protein Phosphatase 2A (CIP2A)/p90 in Cancer. Front Genet 2023; 14:1110656. [PMID: 36911405 PMCID: PMC9998691 DOI: 10.3389/fgene.2023.1110656] [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: 11/29/2022] [Accepted: 02/03/2023] [Indexed: 03/14/2023] Open
Abstract
Cancerous inhibitor of protein phosphatase 2A (CIP2A), initially reported as a tumor-associated antigen (known as p90), is highly expressed in most solid and hematological tumors. The interaction of CIP2A/p90, protein phosphatase 2A (PP2A), and c-Myc can hinder the function of PP2A toward c-Myc S62 induction, thus stabilizing c-Myc protein, which represents a potential role of CIP2A/p90 in tumorigeneses such as cell proliferation, invasion, and migration, as well as cancer drug resistance. The signaling pathways and regulation networks of CIP2A/p90 are complex and not yet fully understood. Many previous studies have also demonstrated that CIP2A/p90 can be used as a potential therapeutic cancer target. In addition, the autoantibody against CIP2A/p90 in sera may be used as a promising biomarker in the diagnosis of certain types of cancer. In this Review, we focus on recent advances relating to CIP2A/p90 and their implications for future research.
Collapse
Affiliation(s)
- Beibei Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Huihui Hu
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Xiaobing Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| |
Collapse
|
47
|
The Pivotal Role of Protein Phosphatase 2A (PP2A) in Brain Tumors. Int J Mol Sci 2022; 23:ijms232415717. [PMID: 36555359 PMCID: PMC9779694 DOI: 10.3390/ijms232415717] [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: 11/25/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022] Open
Abstract
Protein phosphatase 2A (PP2A) is a highly complex heterotrimeric Ser/Thr phosphatase that regulates many cellular processes. PP2A is dysregulated in several human diseases, including oncological pathology; interestingly, PP2A appears to be essential for controlling cell growth and may be involved in cancer development. The role of PP2A as a tumor suppressor has been extensively studied and reviewed. To leverage the potential clinical utility of combination PP2A inhibition and radiotherapy treatment, it is vital that novel highly specific PP2A inhibitors be developed. In this review, the existing literature on the role of PP2A in brain tumors, especially in gliomas and glioblastoma (GBM), was analyzed. Interestingly, the review focused on the role of PP2A inhibitors, focusing on CIP2A inhibition, as CIP2A participated in tumor cell growth by stimulating cell-renewal survival, cellular proliferation, evasion of senescence and inhibition of apoptosis. This review suggested CIP2A inhibition as a promising strategy in oncology target therapy.
Collapse
|
48
|
ERK-mediated NELF-A phosphorylation promotes transcription elongation of immediate-early genes by releasing promoter-proximal pausing of RNA polymerase II. Nat Commun 2022; 13:7476. [PMID: 36463234 PMCID: PMC9719515 DOI: 10.1038/s41467-022-35230-4] [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: 07/09/2021] [Accepted: 11/22/2022] [Indexed: 12/05/2022] Open
Abstract
Growth factor-induced, ERK-mediated induction of immediate-early genes (IEGs) is crucial for cell growth and tumorigenesis. Although IEG expression is mainly regulated at the level of transcription elongation by RNA polymerase-II (Pol-II) promoter-proximal pausing and its release, the role of ERK in this process remains unknown. Here, we identified negative elongation factor (NELF)-A as an ERK substrate. Upon growth factor stimulation, ERK phosphorylates NELF-A, which dissociates NELF from paused Pol-II at the promoter-proximal regions of IEGs, allowing Pol-II to resume elongation and produce full-length transcripts. Furthermore, we found that in cancer cells, PP2A efficiently dephosphorylates NELF-A, thereby preventing aberrant IEG expression induced by ERK-activating oncogenes. However, when PP2A inhibitor proteins are overexpressed, as is frequently observed in cancers, decreased PP2A activity combined with oncogene-mediated ERK activation conspire to induce NELF-A phosphorylation and IEG upregulation, resulting in tumor progression. Our data delineate previously unexplored roles of ERK and PP2A inhibitor proteins in carcinogenesis.
Collapse
|
49
|
Gao S, Shan L, Zhang M, Wang Y, Zhan X, Yin Y, Jiang Z, Tao X, Li X, Ye M, Liu Y. Inhibition of PP2A by LB100 sensitizes bladder cancer cells to chemotherapy by inducing p21 degradation. Cell Oncol 2022; 45:1203-1215. [PMID: 36136269 DOI: 10.1007/s13402-022-00710-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2022] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Bladder carcinoma (BLCA) is the most common urinary tract malignancy and exhibits a poor response to chemotherapy. Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase involved in a wide variety of regulatory cellular processes, including apoptosis and the DNA-damage response (DDR). LB100, a small molecule inhibitor of PP2A, has been shown to act as a chemo-sensitizer in multiple types of cancer. However, the anti-tumor effect and mode of action of LB100 in BLCA have yet to be identified. METHODS In vitro and in vivo experiments were performed to assess the anti-tumor effect of LB100 alone or in combination with gemcitabine. Mass spectrometry (MS)-based phosphoproteomics analysis was used to identify the downstream substrates of PP2A and to explore the mechanism underlying LB100-induced DNA damage and apoptosis. In addition, we established a chemo-resistant BLCA cell line (RT-112-R) by prolonged drug exposure and determined the effect of LB100 in enhancing genotoxicity in BLCA cell lines and xenograft mouse models. RESULTS We found that LB100 is sufficient to induce an anti-tumor response in BLCA cells by inducing DNA damage and apoptosis both in vitro and in vivo. Furthermore, we found that PP2A potentially dephosphorylates p-p21-ser130 to stabilize p21. Inhibition of PP2A by LB100 increased the level of p-p21-ser130, subsequently leading to a reduction in p21 level in a dose-dependent manner. In addition, we found that treatment of LB100 abrogated the G1/S cell cycle checkpoint, resulting in increased phosphorylation of γH2AX in BLCA cells. Moreover, LB100 enhanced genotoxicity in chemo-resistant BLCA cells by inducing DNA damage and apoptosis in vitro and in vivo. CONCLUSION Our findings indicate that PP2A may serve as a potential therapeutic target in BLCA through regulating p21 stability.
Collapse
Affiliation(s)
- Song Gao
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Liping Shan
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Mo Zhang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yan Wang
- (CAS) Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, China Academy of Sciences, Dalian, 116023, China
| | - Xi Zhan
- (CAS) Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, China Academy of Sciences, Dalian, 116023, China
| | - Yalei Yin
- (CAS) Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, China Academy of Sciences, Dalian, 116023, China
| | - Zhonghao Jiang
- Department of Hepatobiliary and Splenic Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xinyi Tao
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.,Innovative Research Center for Integrated Cancer Omics, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xinyu Li
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, 110004, China. .,Innovative Research Center for Integrated Cancer Omics, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Mingliang Ye
- (CAS) Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, China Academy of Sciences, Dalian, 116023, China.
| | - Yang Liu
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, 110004, China. .,Innovative Research Center for Integrated Cancer Omics, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
50
|
Haanen TJ, O'Connor CM, Narla G. Biased holoenzyme assembly of protein phosphatase 2A (PP2A): From cancer to small molecules. J Biol Chem 2022; 298:102656. [PMID: 36328247 PMCID: PMC9707111 DOI: 10.1016/j.jbc.2022.102656] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Protein phosphatase 2A (PP2A) is a family of serine threonine phosphatases responsible for regulating protein phosphorylation, thus opposing the activity of cellular kinases. PP2A is composed of a catalytic subunit (PP2A Cα/β) and scaffolding subunit (PP2A Aα/β) and various substrate-directing B regulatory subunits. PP2A biogenesis is regulated at multiple levels. For example, the sequestration of the free catalytic subunit during the process of biogenesis avoids promiscuous phosphatase activity. Posttranslational modifications of PP2A C direct PP2A heterotrimeric formation. Additionally, PP2A functions as a haploinsufficient tumor suppressor, where attenuated PP2A enzymatic activity creates a permissive environment for oncogenic transformation. Recent work studying PP2A in cancer showed that its role in tumorigenesis is more nuanced, with some holoenzymes being tumor suppressive, while others are required for oncogenic transformation. In cancer biology, PP2A function is modulated through various mechanisms including the displacement of specific B regulatory subunits by DNA tumor viral antigens, by recurrent mutations, and through loss of carboxymethyl-sensitive heterotrimeric complexes. In aggregate, these alterations bias PP2A activity away from its tumor suppressive functions and toward oncogenic ones. From a therapeutic perspective, molecular glues and disruptors present opportunities for both the selective stabilization of tumor-suppressive holoenzymes and disruption of holoenzymes that are pro-oncogenic. Collectively, these approaches represent an attractive cancer therapy for a wide range of tumor types. This review will discuss the mechanisms by which PP2A holoenzyme formation is dysregulated in cancer and the current therapies that are aimed at biasing heterotrimer formation of PP2A for the treatment of cancer.
Collapse
Affiliation(s)
- Terrance J Haanen
- Division of Genetic Medicine, Department of Internal Medicine, The University of Michigan, Ann Arbor, Michigan, USA
| | - Caitlin M O'Connor
- Division of Genetic Medicine, Department of Internal Medicine, The University of Michigan, Ann Arbor, Michigan, USA
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, The University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|