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
|
Chien MH, Hung WY, Lai TC, Tsai CH, Lee KL, Hsieh FK, Lee WJ, Chang JH. The off‑target effect of loratadine triggers autophagy‑mediated apoptosis in lung adenocarcinoma cells by deactivating JNK, p38, and STAT3 signaling through both PP2A‑dependent and independent pathways. Int J Mol Med 2025; 55:54. [PMID: 39886963 PMCID: PMC11819771 DOI: 10.3892/ijmm.2025.5495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/03/2024] [Indexed: 02/01/2025] Open
Abstract
Lung adenocarcinoma (LUAD) is a typical inflammation‑associated cancer, and anti‑inflammatory medications can be valuable in cancer therapy. Loratadine, a histamine receptor H1 (HRH1) antagonist, shows both anti‑inflammatory and anticancer properties. The present study aimed to evaluate impacts of loratadine on LUAD cells as well as in a LUAD xenograft mouse model, and explore underlying mechanisms. Mechanistic investigations were conducted through using western blotting, flow cytometry, immunohistochemistry, acridine orange staining, TUNEL assays, and in silico analyses of loratadine‑modulated genes in LUAD specimens. It was observed that loratadine inhibited LUAD cell proliferation and colony formation by inducing autophagy‑mediated apoptotic cell death independently of HRH1. In a LUAD xenograft model, loratadine decreased tumor proliferation and angiogenesis while enhancing autophagy and apoptosis. Mechanistically, loratadine induced protein phosphatase 2A (PP2A) activation to deactivate c‑Jun N‑terminal kinase (JNK)1/2 and p38 in H23 and PC9 LUAD cells. Additionally, loratadine inhibited signal transducer and activator of transcription 3 (STAT3) activation via a PP2A‑independent pathway. Furthermore, the combination of loratadine with inhibitors for JNK, p38 and STAT3 all enhanced proliferation inhibition of loratadine alone in both cell lines. In the clinic, patients with LUAD expressing high PP2A had favorable prognoses. The present study suggests that loratadine can be used as a PP2A activator for LUAD treatment, and the combination of repurposing loratadine with inhibitors of STAT3, JNK and p38 would be an effectively strategy for inhibiting LUAD growth.
Collapse
Affiliation(s)
- Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan, R.O.C
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan, R.O.C
| | - Wen-Yueh Hung
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
| | - Tsung-Ching Lai
- Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan, R.O.C
- Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan, R.O.C
| | - Ching Han Tsai
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
| | - Kai-Ling Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan, R.O.C
| | - Feng-Koo Hsieh
- The Genome Engineering and Stem Cell Center, School of Medicine, Washington University, St. Louis, MO 63105, USA
| | - Wei-Jiunn Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan, R.O.C
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
| | - Jer-Hwa Chang
- Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan, R.O.C
- Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan, R.O.C
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
| |
Collapse
|
3
|
Zhou X, Xu Q, Hu X, Klenotic PA, Valdivia A, Leshnower BG, Dong N, Narla G, Lin Z. PP2A Attenuates Thoracic Aneurysm and Dissection in Mouse Models of Marfan Syndrome. Hypertension 2025; 82:665-679. [PMID: 39878024 PMCID: PMC11922656 DOI: 10.1161/hypertensionaha.124.23494] [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: 06/10/2024] [Accepted: 01/10/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Recent studies show that hyperactivation of mTOR (mammalian target of rapamycin) signaling plays a causal role in the development of thoracic aortic aneurysm and dissection. Modulation of PP2A (protein phosphatase 2A) activity has been shown to be of significant therapeutic value. In light of the effects that PP2A can exert on the mTOR pathway, we hypothesized that PP2A activation by small-molecule activators of PP2A could mitigate AA progression in Marfan syndrome (MFS). METHODS Two distinct mouse models of MFS underwent daily oral administration of small-molecule activators of the PP2A compound DT-061 to assess its therapeutic potential. Echocardiography was performed to monitor the growth of the aortic root and ascending aorta. Histological evaluation was performed to assess alterations in the vascular wall. RNA-sequencing, Western blot, and immunostaining were performed to decipher the underlying mechanisms by which DT-061 suppresses AA progression. RESULTS PP2A activity decreased, while mTOR activity increased in both human and mouse aortas with MFS. Concordantly, oral administration of DT-061 increased PP2A activation, reducing aortic expansion in Marfan mice. DT-061 treatment also mitigated medial hypertrophy, elastin breakdown, and extracellular matrix deterioration in the ascending aorta, along with decreased metalloproteinase activities. Mechanistic studies suggest that DT-061 suppresses mTOR signaling and smooth muscle cell dedifferentiation, contributing to its effects on thoracic aortic aneurysm and dissection progression. CONCLUSIONS These studies demonstrate a pathological role of PP2A activity loss in the cause of MFS and implicate that activation of PP2A may serve as a novel therapeutic strategy to limit MFS progression, including aortic aneurysm formation.
Collapse
Affiliation(s)
- Xianming Zhou
- Cardiology Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Xu
- Cardiology Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Cardiovascular Surgery, Xiangya Hospital of Central South University, Changsha, China
| | - Xingjian Hu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Philip A. Klenotic
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Alejandra Valdivia
- Cardiology Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Bradley G Leshnower
- Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Zhiyong Lin
- Cardiology Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
4
|
Lum MA, Jonas KA, Parmar S, Black AR, O’Connor CM, Dobersch S, Yamamoto N, Robertson TM, Schutter A, Giambi M, Avelar RA, DiFeo A, Woods NT, Kugel S, Narla G, Black JD. Small-molecule modulators of B56-PP2A restore 4E-BP function to suppress eIF4E-dependent translation in cancer cells. J Clin Invest 2025; 135:e176093. [PMID: 39869680 PMCID: PMC11827888 DOI: 10.1172/jci176093] [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: 09/25/2023] [Accepted: 12/18/2024] [Indexed: 01/29/2025] Open
Abstract
Dysregulated eIF4E-dependent translation is a central driver of tumorigenesis and therapy resistance. eIF4E-binding proteins (4E-BP1/2/3) are major negative regulators of eIF4E-dependent translation that are inactivated in tumors through inhibitory phosphorylation or downregulation. Previous studies have linked PP2A phosphatase(s) to activation of 4E-BP1. Here, we leveraged biased small-molecule activators of PP2A (SMAPs) to explore the role of B56-PP2A(s) in 4E-BP regulation and the potential of B56-PP2A activation for restoring translational control in tumors. SMAP treatment promoted PP2A-dependent hypophosphorylation of 4E-BP1/2, supporting a role for B56-PP2As (e.g., B56α-PP2A) as 4E-BP phosphatases. Unexpectedly, SMAPs induced transcriptional upregulation of 4E-BP1 through a B56-PP2A→TFE3/TFEB→ATF4 axis. Cap-binding and coimmunoprecipitation assays showed that B56-PP2A(s) activation blocks assembly of the eIF4F translation initiation complex, and cap-dependent translation assays confirmed the translation-inhibitory effects of SMAPs. Thus, B56-PP2A(s) orchestrate a translation-repressive program involving transcriptional induction and activation of 4E-BP1. Notably, SMAPs promoted 4E-BP1-dependent apoptosis in tumor cells and potentiated 4E-BP1 function in the presence of ERK or mTOR inhibitors, agents that rely on inhibition of eIF4E-dependent translation for antitumor activity. These findings, combined with the ability of SMAPs to regulate 4E-BP1 in vivo, highlight the potential of PP2A activators for cancer therapy and overcoming therapy resistance.
Collapse
Affiliation(s)
- Michelle A. Lum
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, Omaha, Nebraska, USA
| | - Kayla A. Jonas
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, Omaha, Nebraska, USA
| | - Shreya Parmar
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, Omaha, Nebraska, USA
| | - Adrian R. Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, Omaha, Nebraska, USA
| | - Caitlin M. O’Connor
- Division of Genetic Medicine, Department of Internal Medicine, and
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Stephanie Dobersch
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Naomi Yamamoto
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Tess M. Robertson
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Aidan Schutter
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Miranda Giambi
- Division of Genetic Medicine, Department of Internal Medicine, and
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Rita A. Avelar
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pathology and
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
| | - Analisa DiFeo
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pathology and
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicholas T. Woods
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, Omaha, Nebraska, USA
| | - Sita Kugel
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, and
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Jennifer D. Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, Omaha, Nebraska, USA
| |
Collapse
|
5
|
A Avelar R, Gupta R, Carvette G, da Veiga Leprevost F, Jasti M, Colina J, Teitel J, Nesvizhskii AI, O'Connor CM, Hatzoglou M, Shenolikar S, Arvan P, Narla G, DiFeo A. Integrated stress response plasticity governs normal cell adaptation to chronic stress via the PP2A-TFE3-ATF4 pathway. Cell Death Differ 2024; 31:1761-1775. [PMID: 39349971 PMCID: PMC11618521 DOI: 10.1038/s41418-024-01378-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 10/09/2024] Open
Abstract
The integrated stress response (ISR) regulates cell fate during conditions of stress by leveraging the cell's capacity to endure sustainable and efficient adaptive stress responses. Protein phosphatase 2A (PP2A) activity modulation has been shown to be successful in achieving both therapeutic efficacy and safety across various cancer models. However, the molecular mechanisms driving its selective antitumor effects remain unclear. Here, we show for the first time that ISR plasticity relies on PP2A activation to regulate drug response and dictate cellular survival under conditions of chronic stress. We demonstrate that genetic and chemical modulation of the PP2A leads to chronic proteolytic stress and triggers an ISR to dictate whether the cell lives or dies. More specifically, we uncovered that the PP2A-TFE3-ATF4 pathway governs ISR cell plasticity during endoplasmic reticular and cellular stress independent of the unfolded protein response. We further show that normal cells reprogram their genetic signatures to undergo ISR-mediated adaptation and homeostatic recovery thereby avoiding toxicity following PP2A-mediated stress. Conversely, oncogenic specific cytotoxicity induced by chemical modulation of PP2A is achieved by activating chronic and irreversible ISR in cancer cells. Our findings propose that a differential response to chemical modulation of PP2A is determined by intrinsic ISR plasticity, providing a novel biological vulnerability to selectively induce cancer cell death and improve targeted therapeutic efficacy.
Collapse
Affiliation(s)
- Rita A Avelar
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
| | - Riya Gupta
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
| | - Grace Carvette
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
| | | | - Medhasri Jasti
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
| | - Jose Colina
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
| | - Jessica Teitel
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
| | - Alexey I Nesvizhskii
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Caitlin M O'Connor
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Maria Hatzoglou
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Shirish Shenolikar
- Duke-NUS Medical School, Singapore, Singapore
- Duke University School of Medicine, Durham, NC, USA
| | - Peter Arvan
- Division of Metabolism Endocrinology and Diabetes, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Goutham Narla
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Analisa DiFeo
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA.
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA.
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
6
|
Tinsley SL, Chianis ERD, Shelley RA, Mall GK, Dhiman A, Baral G, Kothandaraman H, Thoma MC, English IA, Daniel CJ, Acosta LCS, Solorio L, Atallah Lanman N, Pasca di Magliano M, Narla G, Dykhuizen EC, Sears RC, Allen-Petersen BL. KRAS-mediated upregulation of CIP2A promotes suppression of PP2A-B56α to initiate pancreatic cancer development. Oncogene 2024; 43:3673-3687. [PMID: 39443726 DOI: 10.1038/s41388-024-03196-w] [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: 12/22/2023] [Revised: 10/04/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
Oncogenic mutations in KRAS are present in ~95% of patients diagnosed with pancreatic ductal adenocarcinoma (PDAC) and are considered the initiating event of pancreatic intraepithelial neoplasia (PanIN) precursor lesions. While it is well established that KRAS mutations drive the activation of oncogenic kinase cascades during pancreatic oncogenesis, the effects of oncogenic KRAS signaling on regulation of phosphatases during this process is not fully appreciated. Protein Phosphatase 2A (PP2A) has been implicated in suppressing KRAS-driven cellular transformation and low PP2A activity is observed in PDAC cells compared to non-transformed cells, suggesting that suppression of PP2A activity is an important step in the overall development of PDAC. In the current study, we demonstrate that KRASG12D induces the expression of an endogenous inhibitor of PP2A activity, Cancerous Inhibitor of PP2A (CIP2A), and phosphorylation of the PP2A substrate, c-MYC. Consistent with these findings, KRASG12D sequestered the specific PP2A subunit responsible for c-MYC degradation, B56α, away from the active PP2A holoenzyme in a CIP2A-dependent manner. During PDAC initiation in vivo, knockout of B56α promoted KRASG12D tumorigenesis by accelerating acinar-to-ductal metaplasia (ADM) and the formation of PanIN lesions. The process of ADM was attenuated ex vivo in response to pharmacological re-activation of PP2A utilizing direct small molecule activators of PP2A (SMAPs). Together, our results suggest that suppression of PP2A-B56α through KRAS signaling can promote the MYC-driven initiation of pancreatic tumorigenesis.
Collapse
Affiliation(s)
- Samantha L Tinsley
- Purdue University Interdisciplinary Life Sciences Program (PULSe), Purdue University, West Lafayette, IN, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Ella Rose D Chianis
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Rebecca A Shelley
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Gaganpreet K Mall
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Alisha Dhiman
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Garima Baral
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Harish Kothandaraman
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Mary C Thoma
- Department of Molecular Medicine and Genetics, Oregon Health and Sciences University, Portland, OR, USA
| | - Isabel A English
- Department of Molecular Medicine and Genetics, Oregon Health and Sciences University, Portland, OR, USA
| | - Colin J Daniel
- Department of Molecular Medicine and Genetics, Oregon Health and Sciences University, Portland, OR, USA
| | | | - Luis Solorio
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Nadia Atallah Lanman
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - Marina Pasca di Magliano
- Department of Internal Medicine, Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
| | - Goutham Narla
- Department of Internal Medicine, Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
| | - Emily C Dykhuizen
- Purdue University Interdisciplinary Life Sciences Program (PULSe), Purdue University, West Lafayette, IN, USA
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Rosalie C Sears
- Department of Molecular Medicine and Genetics, Oregon Health and Sciences University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Sciences University, Portland, OR, USA
| | - Brittany L Allen-Petersen
- Purdue University Interdisciplinary Life Sciences Program (PULSe), Purdue University, West Lafayette, IN, USA.
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
7
|
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
|
8
|
Johnson H, Singh A, Raza A, Sha CM, Wang J, Gowda K, Shen Z, Nair H, Li C, Dokholyan NV, Narayan S, Sharma AK. Identification of a Novel Protein Phosphatase 2A Activator, PPA24, as a Potential Therapeutic for FOLFOX-Resistant Colorectal Cancer. J Med Chem 2024; 67:18070-18089. [PMID: 39004939 DOI: 10.1021/acs.jmedchem.4c01077] [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: 07/16/2024]
Abstract
A series of compounds were designed utilizing molecular modeling and fragment-based design based upon the known protein phosphatase 2A (PP2A) activators, NSC49L and iHAP1, and evaluated for their ability to inhibit the viability of colorectal cancer (CRC) and folinic acid, 5-fluorouracil, and oxaliplatin (FOLFOX)-resistant CRC cells. PPA24 (19a) was identified as the most cytotoxic compound with IC50 values in the range of 2.36-6.75 μM in CRC and FOLFOX-resistant CRC cell lines. It stimulated PP2A activity to a greater extent, displayed lower binding energies through molecular docking, and showed higher binding affinity through surface plasmon resonance for PP2A catalytic subunit α than the known PP2A activators. PPA24 dose-dependently induced apoptosis and oxidative stress, decreased the level of c-Myc expression, and synergistically potentiated cytotoxicity when combined with gemcitabine and cisplatin. Furthermore, a PPA24-encapsulated nanoformulation significantly inhibited the growth of CRC xenografts without systemic toxicities. Together, these results signify the potential of PPA24 as a novel PP2A activator and a prospective therapeutic for CRC and FOLFOX-resistant CRC.
Collapse
Affiliation(s)
- Hannah Johnson
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Amandeep Singh
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Asif Raza
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Congzhou M Sha
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Jian Wang
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Krishne Gowda
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Zhihang Shen
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
| | - Haritha Nair
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, 1200 Newell Drive, Gainesville, Florida 32610, United States
| | - Chenglong Li
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
| | - Nikolay V Dokholyan
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Satya Narayan
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, 1200 Newell Drive, Gainesville, Florida 32610, United States
| | - Arun K Sharma
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| |
Collapse
|
9
|
Avital-Cohen N, Chapnik N, Froy O. Resveratrol Induces Myotube Development by Altering Circadian Metabolism via the SIRT1-AMPK-PP2A Axis. Cells 2024; 13:1069. [PMID: 38920697 PMCID: PMC11201382 DOI: 10.3390/cells13121069] [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: 05/22/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
Resveratrol is a polyphenol known to have metabolic as well as circadian effects. However, there is little information regarding the metabolic and circadian effect of resveratrol on muscle cells. We sought to investigate the metabolic impact of resveratrol throughout the circadian cycle to clarify the associated signaling pathways. C2C12 myotubes were incubated with resveratrol in the presence of increasing concentrations of glucose, and metabolic and clock proteins were measured for 24 h. Resveratrol led to SIRT1, AMPK and PP2A activation. Myotubes treated with increasing glucose concentrations showed higher activation of the mTOR signaling pathway. However, resveratrol did not activate the mTOR signaling pathway, except for P70S6K and S6. In accordance with the reduced mTOR activity, resveratrol led to advanced circadian rhythms and reduced levels of pBMAL1 and CRY1. Resveratrol increased myogenin expression and advanced its rhythms. In conclusion, resveratrol activates the SIRT1-AMPK-PP2A axis, advances circadian rhythms and induces muscle development.
Collapse
Affiliation(s)
| | | | - Oren Froy
- Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel; (N.A.-C.); (N.C.)
| |
Collapse
|
10
|
Coutinho LL, Femino EL, Gonzalez AL, Moffat RL, Heinz WF, Cheng RYS, Lockett SJ, Rangel MC, Ridnour LA, Wink DA. NOS2 and COX-2 Co-Expression Promotes Cancer Progression: A Potential Target for Developing Agents to Prevent or Treat Highly Aggressive Breast Cancer. Int J Mol Sci 2024; 25:6103. [PMID: 38892290 PMCID: PMC11173351 DOI: 10.3390/ijms25116103] [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: 02/28/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Nitric oxide (NO) and reactive nitrogen species (RNS) exert profound biological impacts dictated by their chemistry. Understanding their spatial distribution is essential for deciphering their roles in diverse biological processes. This review establishes a framework for the chemical biology of NO and RNS, exploring their dynamic reactions within the context of cancer. Concentration-dependent signaling reveals distinctive processes in cancer, with three levels of NO influencing oncogenic properties. In this context, NO plays a crucial role in cancer cell proliferation, metastasis, chemotherapy resistance, and immune suppression. Increased NOS2 expression correlates with poor survival across different tumors, including breast cancer. Additionally, NOS2 can crosstalk with the proinflammatory enzyme cyclooxygenase-2 (COX-2) to promote cancer progression. NOS2 and COX-2 co-expression establishes a positive feed-forward loop, driving immunosuppression and metastasis in estrogen receptor-negative (ER-) breast cancer. Spatial evaluation of NOS2 and COX-2 reveals orthogonal expression, suggesting the unique roles of these niches in the tumor microenvironment (TME). NOS2 and COX2 niche formation requires IFN-γ and cytokine-releasing cells. These niches contribute to poor clinical outcomes, emphasizing their role in cancer progression. Strategies to target these markers include direct inhibition, involving pan-inhibitors and selective inhibitors, as well as indirect approaches targeting their induction or downstream effectors. Compounds from cruciferous vegetables are potential candidates for NOS2 and COX-2 inhibition offering therapeutic applications. Thus, understanding the chemical biology of NO and RNS, their spatial distribution, and their implications in cancer progression provides valuable insights for developing targeted therapies and preventive strategies.
Collapse
Affiliation(s)
- Leandro L. Coutinho
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA; (L.L.C.); (E.L.F.); (A.L.G.); (R.Y.S.C.)
- Center for Translational Research in Oncology, ICESP/HC, Faculdade de Medicina da Universidade de São Paulo and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, SP, Brazil;
| | - Elise L. Femino
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA; (L.L.C.); (E.L.F.); (A.L.G.); (R.Y.S.C.)
| | - Ana L. Gonzalez
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA; (L.L.C.); (E.L.F.); (A.L.G.); (R.Y.S.C.)
| | - Rebecca L. Moffat
- Optical Microscopy and Analysis Laboratory, Office of Science and Technology Resources, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA;
| | - William F. Heinz
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (W.F.H.); (S.J.L.)
| | - Robert Y. S. Cheng
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA; (L.L.C.); (E.L.F.); (A.L.G.); (R.Y.S.C.)
| | - Stephen J. Lockett
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (W.F.H.); (S.J.L.)
| | - M. Cristina Rangel
- Center for Translational Research in Oncology, ICESP/HC, Faculdade de Medicina da Universidade de São Paulo and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, SP, Brazil;
| | - Lisa A. Ridnour
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA; (L.L.C.); (E.L.F.); (A.L.G.); (R.Y.S.C.)
| | - David A. Wink
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA; (L.L.C.); (E.L.F.); (A.L.G.); (R.Y.S.C.)
| |
Collapse
|
11
|
Banerjee A, Mathew S, Naqvi MM, Yilmaz SZ, Zacharopoulou M, Doruker P, Kumita JR, Yang SH, Gur M, Itzhaki LS, Gordon R, Bahar I. Influence of point mutations on PR65 conformational adaptability: Insights from molecular simulations and nanoaperture optical tweezers. SCIENCE ADVANCES 2024; 10:eadn2208. [PMID: 38820156 PMCID: PMC11141623 DOI: 10.1126/sciadv.adn2208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/29/2024] [Indexed: 06/02/2024]
Abstract
PR65 is the HEAT repeat scaffold subunit of the heterotrimeric protein phosphatase 2A (PP2A) and an archetypal tandem repeat protein. Its conformational mechanics plays a crucial role in PP2A function by opening/closing substrate binding/catalysis interface. Using in silico saturation mutagenesis, we identified PR65 "hinge" residues whose substitutions could alter its conformational adaptability and thereby PP2A function, and selected six mutations that were verified to be expressed and soluble. Molecular simulations and nanoaperture optical tweezers revealed consistent results on the specific effects of the mutations on the structure and dynamics of PR65. Two mutants observed in simulations to stabilize extended/open conformations exhibited higher corner frequencies and lower translational scattering in experiments, indicating a shift toward extended conformations, whereas another displayed the opposite features, confirmed by both simulations and experiments. The study highlights the power of single-molecule nanoaperture-based tweezers integrated with in silico approaches for exploring the effect of mutations on protein structure and dynamics.
Collapse
Affiliation(s)
- Anupam Banerjee
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Samuel Mathew
- Department of Electrical and Computer Engineering, University of Victoria, Victoria V8P 5C2, Canada
| | - Mohsin M. Naqvi
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Sema Z. Yilmaz
- Department of Mechanical Engineering, Istanbul Technical University, 34437 Istanbul, Turkey
| | - Maria Zacharopoulou
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Pemra Doruker
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Janet R. Kumita
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Shang-Hua Yang
- Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Mert Gur
- Department of Mechanical Engineering, Istanbul Technical University, 34437 Istanbul, Turkey
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Laura S. Itzhaki
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Reuven Gordon
- Department of Electrical and Computer Engineering, University of Victoria, Victoria V8P 5C2, Canada
| | - Ivet Bahar
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA
- Department of Biochemistry and Cell Biology, School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
12
|
Tinsley SL, Shelley RA, Mall GK, Chianis ERD, Dhiman A, Baral G, Kothandaraman H, Thoma MC, Daniel CJ, Lanman NA, di Magliano MP, Narla G, Solorio L, Dykhuizen EC, Sears RC, Allen-Petersen BL. KRAS-mediated upregulation of CIP2A promotes suppression of PP2A-B56α to initiate pancreatic cancer development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.01.547283. [PMID: 38826439 PMCID: PMC11142131 DOI: 10.1101/2023.07.01.547283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Oncogenic mutations in KRAS are present in approximately 95% of patients diagnosed with pancreatic ductal adenocarcinoma (PDAC) and are considered the initiating event of pancreatic intraepithelial neoplasia (PanIN) precursor lesions. While it is well established that KRAS mutations drive the activation of oncogenic kinase cascades during pancreatic oncogenesis, the effects of oncogenic KRAS signaling on regulation of phosphatases during this process is not fully appreciated. Protein Phosphatase 2A (PP2A) has been implicated in suppressing KRAS-driven cellular transformation. However, low PP2A activity is observed in PDAC cells compared to non-transformed cells, suggesting that suppression of PP2A activity is an important step in the overall development of PDAC. In the current study, we demonstrate that KRASG12D induces the expression of both an endogenous inhibitor of PP2A activity, Cancerous Inhibitor of PP2A (CIP2A), and the PP2A substrate, c-MYC. Consistent with these findings, KRASG12D sequestered the specific PP2A subunit responsible for c-MYC degradation, B56α, away from the active PP2A holoenzyme in a CIP2A-dependent manner. During PDAC initiation in vivo, knockout of B56α promoted KRASG12D tumorigenesis by accelerating acinar-to-ductal metaplasia (ADM) and the formation of PanIN lesions. The process of ADM was attenuated ex vivo in response to pharmacological re-activation of PP2A utilizing direct small molecule activators of PP2A (SMAPs). Together, our results suggest that suppression of PP2A-B56α through KRAS signaling can promote the MYC-driven initiation of pancreatic tumorigenesis.
Collapse
Affiliation(s)
- Samantha L. Tinsley
- Purdue University Interdisciplinary Life Sciences Program (PULSe), Purdue University, West Lafayette, IN, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN USA
| | - Rebecca A. Shelley
- Department of Biological Sciences, Purdue University, West Lafayette, IN USA
| | - Gaganpreet K. Mall
- Department of Biological Sciences, Purdue University, West Lafayette, IN USA
| | | | - Alisha Dhiman
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - Garima Baral
- Department of Biological Sciences, Purdue University, West Lafayette, IN USA
| | - Harish Kothandaraman
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Mary C. Thoma
- Department of Molecular Medicine and Genetics, Oregon Health and Sciences University, Portland, Oregon, USA
| | - Colin J. Daniel
- Department of Molecular Medicine and Genetics, Oregon Health and Sciences University, Portland, Oregon, USA
| | - Nadia Atallah Lanman
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | | | - Goutham Narla
- University of Michigan School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Emily C. Dykhuizen
- Purdue University Interdisciplinary Life Sciences Program (PULSe), Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Rosalie C. Sears
- Department of Molecular Medicine and Genetics, Oregon Health and Sciences University, Portland, Oregon, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Sciences University, Portland, Oregon, USA
| | - Brittany L. Allen-Petersen
- Purdue University Interdisciplinary Life Sciences Program (PULSe), Purdue University, West Lafayette, IN, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN USA
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
13
|
Luk IS, Bridgwater CM, Yu A, Boila LD, Yáñez-Bartolomé M, Lampano AE, Hulahan TS, Boukhali M, Kathiresan M, Macarulla T, Kenerson HL, Yamamoto N, Sokolov D, Engstrom IA, Sullivan LB, Lampe PD, Cooper JA, Yeung RS, Tian TV, Haas W, Saha SK, Kugel S. SRC inhibition enables formation of a growth suppressive MAGI1-PP2A complex in isocitrate dehydrogenase-mutant cholangiocarcinoma. Sci Transl Med 2024; 16:eadj7685. [PMID: 38748774 PMCID: PMC11218711 DOI: 10.1126/scitranslmed.adj7685] [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: 07/15/2023] [Accepted: 04/25/2024] [Indexed: 07/04/2024]
Abstract
Intrahepatic cholangiocarcinoma (ICC) is an aggressive bile duct malignancy that frequently exhibits isocitrate dehydrogenase (IDH1/IDH2) mutations. Mutant IDH (IDHm) ICC is dependent on SRC kinase for growth and survival and is hypersensitive to inhibition by dasatinib, but the molecular mechanism underlying this sensitivity is unclear. We found that dasatinib reduced p70 S6 kinase (S6K) and ribosomal protein S6 (S6), leading to substantial reductions in cell size and de novo protein synthesis. Using an unbiased phosphoproteomic screen, we identified membrane-associated guanylate kinase, WW, and PDZ domain containing 1 (MAGI1) as an SRC substrate in IDHm ICC. Biochemical and functional assays further showed that SRC inhibits a latent tumor-suppressing function of the MAGI1-protein phosphatase 2A (PP2A) complex to activate S6K/S6 signaling in IDHm ICC. Inhibiting SRC led to activation and increased access of PP2A to dephosphorylate S6K, resulting in cell death. Evidence from patient tissue and cell line models revealed that both intrinsic and extrinsic resistance to dasatinib is due to increased phospho-S6 (pS6). To block pS6, we paired dasatinib with the S6K/AKT inhibitor M2698, which led to a marked reduction in pS6 in IDHm ICC cell lines and patient-derived organoids in vitro and substantial growth inhibition in ICC patient-derived xenografts in vivo. Together, these results elucidated the mechanism of action of dasatinib in IDHm ICC, revealed a signaling complex regulating S6K phosphorylation independent of mTOR, suggested markers for dasatinib sensitivity, and described a combination therapy for IDHm ICC that may be actionable in the clinic.
Collapse
Affiliation(s)
- Iris S. Luk
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | | | - Angela Yu
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Liberalis D. Boila
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Mariana Yáñez-Bartolomé
- Preclinical and Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Aaron E. Lampano
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Taylor S. Hulahan
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Myriam Boukhali
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| | - Meena Kathiresan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| | - Teresa Macarulla
- Preclinical and Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
- Gastrointestinal and Endocrine Tumor Unit, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - Heidi L. Kenerson
- Department of Surgery, University of Washington, Seattle, WA 98195, USA
| | - Naomi Yamamoto
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - David Sokolov
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Ian A. Engstrom
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Lucas B. Sullivan
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Paul D. Lampe
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Jonathan A. Cooper
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Raymond S. Yeung
- Department of Surgery, University of Washington, Seattle, WA 98195, USA
| | - Tian V. Tian
- Preclinical and Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Wilhelm Haas
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA
| | - Supriya K. Saha
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Sita Kugel
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| |
Collapse
|
14
|
Zhou X, Xu Q, Li W, Dong N, Stomberski C, Narla G, Lin Z. Protein Phosphatase 2A Activation Promotes Heart Transplant Acceptance in Mice. Transplantation 2024; 108:e36-e48. [PMID: 38126420 PMCID: PMC10922415 DOI: 10.1097/tp.0000000000004832] [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] [Indexed: 12/23/2023]
Abstract
BACKGROUND Although heart transplantation is the definitive treatment for heart failure in eligible patients, both acute and chronic transplant rejection frequently occur. Protein phosphatase 2A (PP2A) activity is critical in maintaining tissue and organ homeostasis. In this study, we evaluated the effect of a novel class of small molecule activators of PP2A (SMAPs) on allograft rejection in a mouse heterotopic heart transplantation model. METHODS Recipient mice were administered with DT-061 (a pharmaceutically optimized SMAP) or vehicle by oral gavage beginning 1 d after transplantation. Histological and immunofluorescence analyses were performed to examine allograft rejection. Regulatory T cells (Treg) from recipient spleens were subjected to flow cytometry and RNA sequencing analysis. Finally, the effect of DT-061 on smooth muscle cells (SMCs) migration and proliferation was assessed. RESULTS DT-061 treatment prolonged cardiac allograft survival. SMAPs effectively suppressed the inflammatory immune response while increasing Treg population in the allografts, findings corroborated by functional analysis of RNA sequencing data derived from Treg of treated splenic tissues. Importantly, SMAPs extended immunosuppressive agent cytotoxic T lymphocyte-associated antigen-4-Ig-induced cardiac transplantation tolerance and allograft survival. SMAPs also strongly mitigated cardiac allograft vasculopathy as evidenced by a marked reduction of neointimal hyperplasia and SMC proliferation. Finally, our in vitro studies implicate suppression of MEK/ERK pathways as a unifying mechanism for the effect of PP2A modulation in Treg and SMCs. CONCLUSIONS PP2A activation prevents cardiac rejection and prolongs allograft survival in a murine model. Our findings highlight the potential of PP2A activation in improving alloengraftment in heart transplantation.
Collapse
Affiliation(s)
- Xianming Zhou
- Cardiology Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Xu
- Cardiology Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Cardiovascular Surgery, Xiangya Hospital of Central South University, Changsha, China
| | - Wangzi Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Colin Stomberski
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Zhiyong Lin
- Cardiology Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
15
|
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
|
16
|
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: 9] [Impact Index Per Article: 4.5] [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
|
17
|
Jayappa KD, Tran B, Gordon VL, Morris C, Saha S, Farrington CC, O’Connor CM, Zawacki KP, Isaac KM, Kester M, Bender TP, Williams ME, Portell CA, Weber MJ, Narla G. PP2A modulation overcomes multidrug resistance in chronic lymphocytic leukemia via mPTP-dependent apoptosis. J Clin Invest 2023; 133:e155938. [PMID: 37166997 PMCID: PMC10313372 DOI: 10.1172/jci155938] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 05/09/2023] [Indexed: 05/12/2023] Open
Abstract
Targeted therapies such as venetoclax (VEN) (Bcl-2 inhibitor) have revolutionized the treatment of chronic lymphocytic leukemia (CLL). We previously reported that persister CLL cells in treated patients overexpress multiple antiapoptotic proteins and display resistance to proapoptotic agents. Here, we demonstrated that multidrug-resistant CLL cells in vivo exhibited apoptosis restriction at a pre-mitochondrial level due to insufficient activation of the Bax and Bak (Bax/Bak) proteins. Co-immunoprecipitation analyses with selective BH domain antagonists revealed that the pleiotropic proapoptotic protein (Bim) was prevented from activating Bax/Bak by "switching" interactions to other upregulated antiapoptotic proteins (Mcl-1, Bcl-xL, Bcl-2). Hence, treatments that bypass Bax/Bak restriction are required to deplete these resistant cells in patients. Protein phosphatase 2A (PP2A) contributes to oncogenesis and treatment resistance. We observed that small-molecule activator of PP2A (SMAP) induced cytotoxicity in multiple cancer cell lines and CLL samples, including multidrug-resistant leukemia and lymphoma cells. The SMAP (DT-061) activated apoptosis in multidrug-resistant CLL cells through induction of mitochondrial permeability transition pores, independent of Bax/Bak. DT-061 inhibited the growth of wild-type and Bax/Bak double-knockout, multidrug-resistant CLL cells in a xenograft mouse model. Collectively, we discovered multidrug-resistant CLL cells in patients and validated a pharmacologically tractable pathway to deplete this reservoir.
Collapse
MESH Headings
- Humans
- Animals
- Mice
- bcl-2-Associated X Protein/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Protein Phosphatase 2/genetics
- Myeloid Cell Leukemia Sequence 1 Protein/metabolism
- Proto-Oncogene Proteins c-bcl-2
- Apoptosis/physiology
- Apoptosis Regulatory Proteins/metabolism
- Drug Resistance, Multiple
Collapse
Affiliation(s)
- Kallesh D. Jayappa
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia (UVA) School of Medicine, Charlottesville, Virginia, USA
- Beirne B. Carter Center for Immunology Research, Charlottesville, Virginia, USA
- Department of Pharmacology, Charlottesville, Virginia, USA
| | - Brian Tran
- Division of Genetic Medicine, Department of Internal Medicine, the University of Michigan, Ann Arbor, Michigan, USA
| | - Vicki L. Gordon
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia (UVA) School of Medicine, Charlottesville, Virginia, USA
- Beirne B. Carter Center for Immunology Research, Charlottesville, Virginia, USA
| | - Christopher Morris
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia (UVA) School of Medicine, Charlottesville, Virginia, USA
- Beirne B. Carter Center for Immunology Research, Charlottesville, Virginia, USA
| | - Shekhar Saha
- Department of Biochemistry and Molecular Genetics, Charlottesville, Virginia, USA
| | - Caroline C. Farrington
- 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
| | - Kaitlin P. Zawacki
- Division of Genetic Medicine, Department of Internal Medicine, the University of Michigan, Ann Arbor, Michigan, USA
| | - Krista M. Isaac
- Division of Hematology/Oncology, UVA School of Medicine, Charlottesville, Virginia, USA
| | - Mark Kester
- Department of Pharmacology, Charlottesville, Virginia, USA
- Department of Biomedical Engineering, UVA, Charlottesville, Virginia, USA
| | - Timothy P. Bender
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia (UVA) School of Medicine, Charlottesville, Virginia, USA
- Beirne B. Carter Center for Immunology Research, Charlottesville, Virginia, USA
| | - Michael E. Williams
- Division of Hematology/Oncology, UVA School of Medicine, Charlottesville, Virginia, USA
- Cancer Center, UVA, Charlottesville, Virginia, USA
| | - Craig A. Portell
- Division of Hematology/Oncology, UVA School of Medicine, Charlottesville, Virginia, USA
- Cancer Center, UVA, Charlottesville, Virginia, USA
| | - Michael J. Weber
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia (UVA) School of Medicine, Charlottesville, Virginia, USA
- Beirne B. Carter Center for Immunology Research, Charlottesville, Virginia, USA
- Cancer Center, UVA, Charlottesville, Virginia, USA
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, the University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
18
|
Bownes LV, Julson JR, Quinn CH, Hutchins SC, Erwin MH, Markert HR, Stewart JE, Mroczek-Musulman E, Aye J, Yoon KJ, Ohlmeyer M, Beierle EA. The Effects of Protein Phosphatase 2A Activation with Novel Tricyclic Sulfonamides on Hepatoblastoma. J Pediatr Surg 2023; 58:1145-1154. [PMID: 36907775 PMCID: PMC10198925 DOI: 10.1016/j.jpedsurg.2023.02.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/10/2023] [Indexed: 02/19/2023]
Abstract
BACKGROUND The tumor suppressor, protein phosphatase 2A (PP2A), is downregulated in hepatoblastoma. We aimed to examine the effects of two novel compounds of the tricyclic sulfonamide class, ATUX-3364 (3364) and ATUX-8385 (8385), designed to activate PP2A without causing immunosuppression, on human hepatoblastoma. METHODS An established human hepatoblastoma cell line, HuH6, and a human hepatoblastoma patient-derived xenograft, COA67, were treated with increasing doses of 3364 or 8385, and viability, proliferation, cell cycle and motility were investigated. Cancer cell stemness was evaluated by real-time PCR and tumorsphere forming ability. Effects on tumor growth were examined using a murine model. RESULTS Treatment with 3364 or 8385 significantly decreased viability, proliferation, cell cycle progression and motility in HuH6 and COA67 cells. Both compounds significantly decreased stemness as demonstrated by decreased abundance of OCT4, NANOG, and SOX2 mRNA. The ability of COA67 to form tumorspheres, another sign of cancer cell stemness, was significantly diminished by 3364 and 8385. Treatment with 3364 resulted in decreased tumor growth in vivo. CONCLUSION Novel PP2A activators, 3364 and 8385, decreased hepatoblastoma proliferation, viability, and cancer cell stemness in vitro. Animals treated with 3364 had decreased tumor growth. These data provide evidence for further investigation of PP2A activating compounds as hepatoblastoma therapeutics.
Collapse
Affiliation(s)
- Laura V Bownes
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Janet R Julson
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Colin H Quinn
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Sara Claire Hutchins
- Division of Hematology/Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Michael H Erwin
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Hooper R Markert
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Jerry E Stewart
- Division of Pediatric Surgery, Department of Surgery, 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
| | - Karina J Yoon
- Department of Pharmacology and Toxicology, 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.
| |
Collapse
|
19
|
Avelar RA, Armstrong AJ, Carvette G, Gupta R, Puleo N, Colina JA, Joseph P, Sobeck AM, O'Connor CM, Raines B, Gandhi A, Dziubinski ML, Ma DS, Resnick K, Singh S, Zanotti K, Nagel C, Waggoner S, Thomas DG, Skala SL, Zhang J, Narla G, DiFeo A. Small-Molecule-Mediated Stabilization of PP2A Modulates the Homologous Recombination Pathway and Potentiates DNA Damage-Induced Cell Death. Mol Cancer Ther 2023; 22:599-615. [PMID: 36788429 PMCID: PMC10157366 DOI: 10.1158/1535-7163.mct-21-0880] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 05/17/2022] [Accepted: 02/09/2023] [Indexed: 02/16/2023]
Abstract
High-grade serous carcinoma (HGSC) is the most common and lethal ovarian cancer subtype. PARP inhibitors (PARPi) have become the mainstay of HGSC-targeted therapy, given that these tumors are driven by a high degree of genomic instability (GI) and homologous recombination (HR) defects. Nonetheless, approximately 30% of patients initially respond to treatment, ultimately relapsing with resistant disease. Thus, despite recent advances in drug development and an increased understanding of genetic alterations driving HGSC progression, mortality has not declined, highlighting the need for novel therapies. Using a small-molecule activator of protein phosphatase 2A (PP2A; SMAP-061), we investigated the mechanism by which PP2A stabilization induces apoptosis in patient-derived HGSC cells and xenograft (PDX) models alone or in combination with PARPi. We uncovered that PP2A genes essential for cellular transformation (B56α, B56γ, and PR72) and basal phosphatase activity (PP2A-A and -C) are heterozygously lost in the majority of HGSC. Moreover, loss of these PP2A genes correlates with worse overall patient survival. We show that SMAP-061-induced stabilization of PP2A inhibits the HR output by targeting RAD51, leading to chronic accumulation of DNA damage and ultimately apoptosis. Furthermore, combination of SMAP-061 and PARPi leads to enhanced apoptosis in both HR-proficient and HR-deficient HGSC cells and PDX models. Our studies identify PP2A as a novel regulator of HR and indicate PP2A modulators as a therapeutic therapy for HGSC. In summary, our findings further emphasize the potential of PP2A modulators to overcome PARPi insensitivity, given that targeting RAD51 presents benefits in overcoming PARPi resistance driven by BRCA1/2 mutation reversions.
Collapse
Affiliation(s)
- Rita A. Avelar
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | | | - Gracie Carvette
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Riya Gupta
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Noah Puleo
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Jose A. Colina
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Peronne Joseph
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Alexander M. Sobeck
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Caitlin M. O'Connor
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Department of Internal Medicine, Genetic Medicine, University of Michigan, Ann Arbor, Michigan
| | - Brynne Raines
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Department of Internal Medicine, Genetic Medicine, University of Michigan, Ann Arbor, Michigan
| | - Agharnan Gandhi
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Michele L. Dziubinski
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Daniel S. Ma
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | | | | | | | | | | | - Daffyd G. Thomas
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | | | - Junran Zhang
- Department of Radiation Oncology, Ohio State University, Columbus, Ohio
| | - Goutham Narla
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Department of Internal Medicine, Genetic Medicine, University of Michigan, Ann Arbor, Michigan
| | - Analisa DiFeo
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
20
|
Hwang S, Ha Y, Koo G, Noh H, Lee A, Kim B, Hong SM, Morgan MJ, Eyun S, Lee D, Roe J, Lee Y, Kim Y. LCK-Mediated RIPK3 Activation Controls Double-Positive Thymocyte Proliferation and Restrains Thymic Lymphoma by Regulating the PP2A-ERK Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2204522. [PMID: 36161785 PMCID: PMC9661840 DOI: 10.1002/advs.202204522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/07/2022] [Indexed: 06/16/2023]
Abstract
Receptor-interacting protein kinase 3 (RIPK3) is the primary regulator of necroptotic cell death. RIPK3 expression is often silenced in various cancer cells, which suggests that it may have tumor suppressor properties. However, the exact mechanism by which RIPK3 negatively regulates cancer development and progression remains unclear. This report indicates that RIPK3 acts as a potent regulator of the homeostatic proliferation of CD4+ CD8+ double-positive (DP) thymocytes. Abnormal proliferation of RIPK3-deficient DP thymocytes occurs independently of the well-known role for RIPK3 in necroptosis (upstream of MLKL activation), and is associated with an incidental thymic mass, likely thymic hyperplasia. In addition, Ripk3-null mice develop increased thymic tumor formation accompanied by reduced host survival in the context of an N-ethyl-N-nitrosourea (ENU)-induced tumor model. Moreover, RIPK3 deficiency in p53-null mice promotes thymic lymphoma development via upregulated extracellular signal-regulated kinase (ERK) signaling, which correlates with markedly reduced survival rates. Mechanistically, lymphocyte-specific protein tyrosine kinase (LCK) activates RIPK3, which in turn leads to increases in the phosphatase activity of protein phosphatase 2 (PP2A), thereby suppressing hyper-activation of ERK in DP thymocytes. Overall, these findings suggest that a RIPK3-PP2A-ERK signaling axis regulates DP thymocyte homeostasis and may provide a potential therapeutic target to improve thymic lymphoma therapies.
Collapse
Affiliation(s)
- Sung‐Min Hwang
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
- Sandra and Edward Meyer Cancer Center and Department of Obstetrics and GynecologyWeill Cornell MedicineNew YorkNY10065USA
| | - Yu‐Jin Ha
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| | - Gi‐Bang Koo
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| | - Hyun‐Jin Noh
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| | - A‐Yeon Lee
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| | - Byeong‐Ju Kim
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| | - Sun Mi Hong
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| | - Michael J. Morgan
- Department of Natural SciencesNortheastern State UniversityTahlequahOK74464USA
| | - Seong‐il Eyun
- Department of Life ScienceChung‐Ang UniversitySeoul06973Republic of Korea
| | - Dakeun Lee
- Department of PathologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| | - Jae‐Seok Roe
- Department of BiochemistryCollege of Life Science and BiotechnologyYonsei UniversitySeoul03722Republic of Korea
| | - Youngsoo Lee
- Institute of Medical ScienceAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| | - You‐Sun Kim
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| |
Collapse
|
21
|
De Falco F, Rompietti C, Sorcini D, Esposito A, Scialdone A, Baldoni S, Del Papa B, Adamo FM, Silva Barcelos EC, Dorillo E, Stella A, Di Ianni M, Screpanti I, Sportoletti P, Rosati E. GSK3β is a critical, druggable component of the network regulating the active NOTCH1 protein and cell viability in CLL. Cell Death Dis 2022; 13:755. [PMID: 36050315 PMCID: PMC9436923 DOI: 10.1038/s41419-022-05178-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 01/21/2023]
Abstract
NOTCH1 alterations have been associated with chronic lymphocytic leukemia (CLL), but the molecular mechanisms underlying NOTCH1 activation in CLL cells are not completely understood. Here, we show that GSK3β downregulates the constitutive levels of the active NOTCH1 intracellular domain (N1-ICD) in CLL cells. Indeed, GSK3β silencing by small interfering RNA increases N1-ICD levels, whereas expression of an active GSK3β mutant reduces them. Additionally, the GSK3β inhibitor SB216763 enhances N1-ICD stability at a concentration at which it also increases CLL cell viability. We also show that N1-ICD is physically associated with GSK3β in CLL cells. SB216763 reduces GSK3β/N1-ICD interactions and the levels of ubiquitinated N1-ICD, indicating a reduction in N1-ICD proteasomal degradation when GSK3β is less active. We then modulated the activity of two upstream regulators of GSK3β and examined the impact on N1-ICD levels and CLL cell viability. Specifically, we inhibited AKT that is a negative regulator of GSK3β and is constitutively active in CLL cells. Furthermore, we activated the protein phosphatase 2 A (PP2A) that is a positive regulator of GSK3β, and has an impaired activity in CLL. Results show that either AKT inhibition or PP2A activation reduce N1-ICD expression and CLL cell viability in vitro, through mechanisms mediated by GSK3β activity. Notably, for PP2A activation, we used the highly specific activator DT-061, that also reduces leukemic burden in peripheral blood, spleen and bone marrow in the Eµ-TCL1 adoptive transfer model of CLL, with a concomitant decrease in N1-ICD expression. Overall, we identify in GSK3β a key component of the network regulating N1-ICD stability in CLL, and in AKT and PP2A new druggable targets for disrupting NOTCH1 signaling with therapeutic potential.
Collapse
Affiliation(s)
- Filomena De Falco
- grid.9027.c0000 0004 1757 3630Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Chiara Rompietti
- grid.9027.c0000 0004 1757 3630Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Daniele Sorcini
- grid.9027.c0000 0004 1757 3630Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Angela Esposito
- grid.9027.c0000 0004 1757 3630Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Annarita Scialdone
- grid.9027.c0000 0004 1757 3630Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Stefano Baldoni
- grid.9027.c0000 0004 1757 3630Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy ,grid.412451.70000 0001 2181 4941Department of Medicine and Sciences of Aging, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Beatrice Del Papa
- grid.9027.c0000 0004 1757 3630Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Francesco Maria Adamo
- grid.9027.c0000 0004 1757 3630Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Estevão Carlos Silva Barcelos
- grid.9027.c0000 0004 1757 3630Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Erica Dorillo
- grid.9027.c0000 0004 1757 3630Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Arianna Stella
- grid.9027.c0000 0004 1757 3630Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Mauro Di Ianni
- grid.412451.70000 0001 2181 4941Department of Medicine and Sciences of Aging, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy ,grid.461844.bDepartment of Oncology and Hematology, Ospedale Civile “Santo Spirito”, ASL Pescara, Pescara, Italy
| | - Isabella Screpanti
- grid.7841.aDepartment of Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy
| | - Paolo Sportoletti
- grid.9027.c0000 0004 1757 3630Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Emanuela Rosati
- grid.9027.c0000 0004 1757 3630Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
22
|
Narayan S, Raza A, Mahmud I, Koo N, Garrett TJ, Law ME, Law BK, Sharma AK. Sensitization of FOLFOX-resistant colorectal cancer cells via the modulation of a novel pathway involving protein phosphatase 2A. iScience 2022; 25:104518. [PMID: 35754740 PMCID: PMC9218363 DOI: 10.1016/j.isci.2022.104518] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 05/16/2022] [Accepted: 05/30/2022] [Indexed: 12/04/2022] Open
Abstract
The treatment of colorectal cancer (CRC) with FOLFOX shows some efficacy, but these tumors quickly develop resistance to this treatment. We have observed increased phosphorylation of AKT1/mTOR/4EBP1 and levels of p21 in FOLFOX-resistant CRC cells. We have identified a small molecule, NSC49L, that stimulates protein phosphatase 2A (PP2A) activity, downregulates the AKT1/mTOR/4EBP1-axis, and inhibits p21 translation. We have provided evidence that NSC49L- and TRAIL-mediated sensitization is synergistically induced in p21-knockdown CRC cells, which is reversed in p21-overexpressing cells. p21 binds with procaspase 3 and prevents the activation of caspase 3. We have shown that TRAIL induces apoptosis through the activation of caspase 3 by NSC49L-mediated downregulation of p21 translation, and thereby cleavage of procaspase 3 into caspase 3. NSC49L does not affect global protein synthesis. These studies provide a mechanistic understanding of NSC49L as a PP2A agonist, and how its combination with TRAIL sensitizes FOLFOX-resistant CRC cells.
Collapse
Affiliation(s)
- Satya Narayan
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| | - Asif Raza
- Department of Pharmacology, Penn State University College of Medicine, Penn State Cancer Institute, Hershey, PA 17033, USA
| | - Iqbal Mahmud
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Nayeong Koo
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| | - Timothy J. Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mary E. Law
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - Brian K. Law
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - Arun K. Sharma
- Department of Pharmacology, Penn State University College of Medicine, Penn State Cancer Institute, Hershey, PA 17033, USA
| |
Collapse
|
23
|
Wang Q, Xu C, Cai R, An W, Yuan H, Xu M. Fbxo45-mediated NP-STEP 46 degradation via K6-linked ubiquitination sustains ERK activity in lung cancer. Mol Oncol 2022; 16:3017-3033. [PMID: 35838331 PMCID: PMC9394119 DOI: 10.1002/1878-0261.13290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/19/2022] [Accepted: 07/13/2022] [Indexed: 11/15/2022] Open
Abstract
Lung cancer is one of the most threatening malignant tumors to human health. Epidermal growth factor receptor (EGFR)‐targeted therapy is a common and essential means for the clinical treatment of lung cancer. However, drug resistance has always affected the therapeutic effect and survival rate in non‐small cell lung cancer (NSCLC). Tumor heterogeneity is a significant reason, yielding various drug resistance mechanisms, such as EGFR‐dependent or ‐independent extracellular signal‐regulated kinase 1 and/or 2 (ERK1/2) activation in NSCLC. To examine whether this aberrant activation of ERK1/2 is related to the loss of function of its specific phosphatase, a series of in vitro and in vivo assays were performed. We found that F‐box/SPRY domain‐containing protein 1 (Fbxo45) induces ubiquitination of NP‐STEP46, an active form of striatal‐enriched protein tyrosine phosphatase, with a K6‐linked poly‐ubiquitin chain. This ubiquitination led to proteasome degradation in the nucleus, which then sustains the aberrant level of phosphorylated‐ERK (pERK) and promotes tumor growth of NSCLC. Fbxo45 silencing can significantly inhibit cell proliferation and tumor growth. Moreover, NSCLC cells with silenced Fbxo45 showed great sensitivity to the EGFR tyrosine kinase inhibitor (TKI) afatinib. Here, we first report this critical pERK maintenance mechanism, which might be independent of the upstream kinase activity in NSCLC. We propose that inhibiting Fbxo45 may combat the issue of drug resistance in NSCLC patients, especially combining with EGFR‐TKI therapy.
Collapse
Affiliation(s)
- Qian Wang
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai 201999, China
| | - Ci Xu
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai 201999, China.,Department of Gastroenterology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai 201999, China
| | - Renjie Cai
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai 201999, China
| | - Weishu An
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai 201999, China
| | - Haihua Yuan
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai 201999, China
| | - Ming Xu
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai 201999, China
| |
Collapse
|
24
|
Manoharan GB, Okutachi S, Abankwa D. Potential of phenothiazines to synergistically block calmodulin and reactivate PP2A in cancer cells. PLoS One 2022; 17:e0268635. [PMID: 35617282 PMCID: PMC9135253 DOI: 10.1371/journal.pone.0268635] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 05/04/2022] [Indexed: 11/19/2022] Open
Abstract
Phenothiazines (PTZ) were developed as inhibitors of monoamine neurotransmitter receptors, notably dopamine receptors. Because of this activity they have been used for decades as antipsychotic drugs. In addition, they possess significant anti-cancer properties and several attempts for their repurposing were made. However, their incompletely understood polypharmacology is challenging. Here we examined the potential of the PTZ fluphenazine (Flu) and its mustard derivative (Flu-M) to synergistically act on two cancer associated targets, calmodulin (CaM) and the tumor suppressor protein phosphatase 2A (PP2A). Both proteins are known to modulate the Ras- and MAPK-pathway, cell viability and features of cancer cell stemness. Consistently, we show that the combination of a CaM inhibitor and the PP2A activator DT-061 synergistically inhibited the 3D-spheroid formation of MDA-MB-231 (K-Ras-G13D), NCI-H358 (K-Ras-G12C) and A375 (B-raf-V600E) cancer cells, and increased apoptosis in MDA-MB-231. We reasoned that these activities remain combined in PTZ, which were the starting point for PP2A activator development, while several PTZ are known CaM inhibitors. We show that both Flu and Flu-M retained CaM inhibitory activity in vitro and in cells, with a higher potency of the mustard derivative in cells. In line with the CaM dependence of Ras plasma membrane organization, the mustard derivative potently reduced the functional membrane organization of oncogenic Ras, while DT-061 had a negligible effect. Like DT-061, both PTZ potently decreased c-MYC levels, a hallmark of PP2A activation. Benchmarking against the KRAS-G12C specific inhibitor AMG-510 in MIA PaCa-2 cells revealed a higher potency of Flu-M than combinations of DT-061 and a CaM inhibitor on MAPK-output and a strong effect on cell proliferation. While our study is limited, our results suggest that improved PTZ derivatives that retain both, their CaM inhibitory and PP2A activating properties, but have lost their neurological side-effects, may be interesting to pursue further as anti-cancer agents.
Collapse
Affiliation(s)
- Ganesh Babu Manoharan
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sunday Okutachi
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Daniel Abankwa
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- * E-mail:
| |
Collapse
|
25
|
Bownes LV, Marayati R, Quinn CH, Beierle AM, Hutchins SC, Julson JR, Erwin MH, Stewart JE, Mroczek-Musulman E, Ohlmeyer M, Aye JM, Yoon KJ, Beierle EA. Pre-Clinical Study Evaluating Novel Protein Phosphatase 2A Activators as Therapeutics for Neuroblastoma. Cancers (Basel) 2022; 14:1952. [PMID: 35454859 PMCID: PMC9026148 DOI: 10.3390/cancers14081952] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/05/2022] [Accepted: 04/08/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Protein phosphatase 2A (PP2A) functions as an inhibitor of cancer cell proliferation, and its tumor suppressor function is attenuated in many cancers. Previous studies utilized FTY720, an immunomodulating compound known to activate PP2A, and demonstrated a decrease in the malignant phenotype in neuroblastoma. We wished to investigate the effects of two novel PP2A activators, ATUX-792 (792) and DBK-1154 (1154). METHODS Long-term passage neuroblastoma cell lines and human neuroblastoma patient-derived xenograft (PDX) cells were used. Cells were treated with 792 or 1154, and viability, proliferation, and motility were examined. The effect on tumor growth was investigated using a murine flank tumor model. RESULTS Treatment with 792 or 1154 resulted in PP2A activation, decreased cell survival, proliferation, and motility in neuroblastoma cells. Immunoblotting revealed a decrease in MYCN protein expression with increasing concentrations of 792 and 1154. Treatment with 792 led to tumor necrosis and decreased tumor growth in vivo. CONCLUSIONS PP2A activation with 792 or 1154 decreased survival, proliferation, and motility of neuroblastoma in vitro and tumor growth in vivo. Both compounds resulted in decreased expression of the oncogenic protein MYCN. These findings indicate a potential therapeutic role for these novel PP2A activators in neuroblastoma.
Collapse
Affiliation(s)
- Laura V. Bownes
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (L.V.B.); (R.M.); (C.H.Q.); (A.M.B.); (J.R.J.); (M.H.E.); (J.E.S.)
| | - Raoud Marayati
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (L.V.B.); (R.M.); (C.H.Q.); (A.M.B.); (J.R.J.); (M.H.E.); (J.E.S.)
| | - Colin H. Quinn
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (L.V.B.); (R.M.); (C.H.Q.); (A.M.B.); (J.R.J.); (M.H.E.); (J.E.S.)
| | - Andee M. Beierle
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (L.V.B.); (R.M.); (C.H.Q.); (A.M.B.); (J.R.J.); (M.H.E.); (J.E.S.)
| | - Sara C. Hutchins
- Division of Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (S.C.H.); (J.M.A.)
| | - Janet R. Julson
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (L.V.B.); (R.M.); (C.H.Q.); (A.M.B.); (J.R.J.); (M.H.E.); (J.E.S.)
| | - Michael H. Erwin
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (L.V.B.); (R.M.); (C.H.Q.); (A.M.B.); (J.R.J.); (M.H.E.); (J.E.S.)
| | - Jerry E. Stewart
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (L.V.B.); (R.M.); (C.H.Q.); (A.M.B.); (J.R.J.); (M.H.E.); (J.E.S.)
| | | | | | - Jamie M. Aye
- Division of Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (S.C.H.); (J.M.A.)
| | - Karina J. Yoon
- Department of Pharmacology and Toxicology, 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; (L.V.B.); (R.M.); (C.H.Q.); (A.M.B.); (J.R.J.); (M.H.E.); (J.E.S.)
| |
Collapse
|
26
|
Chatam O, Chapnik N, Froy O. Resveratrol Induces the Fasting State and Alters Circadian Metabolism in Hepatocytes. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2022; 77:128-134. [PMID: 35178649 DOI: 10.1007/s11130-022-00954-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/03/2022] [Indexed: 06/14/2023]
Abstract
Resveratrol is a nutritional substance that has both metabolic and circadian effects. While some studies indicate a correlation between resveratrol and reduced gluconeogenesis, others propose the opposite. Our aim was to study the metabolic effect of resveratrol around the circadian clock in order to determine more accurately the hepatic signaling pathways involved. AML-12 hepatocytes were treated with resveratrol and clock and metabolic markers were measured around the clock. Resveratrol-treated AML-12 hepatocytes showed reduced ratio of the following key metabolic factors: phosphorylated PP2A to total PP2A (pPP2A/PP2A), pAKT/AKT, pFOXO1/FOXO1 and pAMPK/AMPK, indicating inhibition of AKT and AMPK, but activation of PP2A and FOXO1. In addition, the levels of phosphorylated mTOR were low after resveratrol treatment. The levels of the key gluconeogenic enzyme phosphoenolpyruvate carboxykinase (PEPCK) were significantly higher after resveratrol treatment. In accordance with the reduced mTOR activity, the ratio of pBMAL1/BMAL1, the clock transcription factor, also decreased. Bmal1 mRNA oscillated robustly in AML-12 hepatocytes, but resveratrol treatment led to a phase advance and a decrease in its amplitude, similarly to the effect on Srebp1c and Pgc1α mRNA. After resveratrol treatment, daily mRNA levels of Bmal1, Sirt1 and Srebp1c were significantly higher. Resveratrol changes the circadian expression of metabolic and clock genes activating the fasting state and inducing the PP2A-FOXO1-PEPCK pathway.
Collapse
Affiliation(s)
- Opal Chatam
- Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 76100, Rehovot, Israel
| | - Nava Chapnik
- Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 76100, Rehovot, Israel
| | - Oren Froy
- Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 76100, Rehovot, Israel.
| |
Collapse
|
27
|
Tinsley SL, Allen-Petersen BL. PP2A and cancer epigenetics: a therapeutic opportunity waiting to happen. NAR Cancer 2022; 4:zcac002. [PMID: 35118387 PMCID: PMC8807117 DOI: 10.1093/narcan/zcac002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/08/2021] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
The epigenetic state of chromatin is altered by regulators which influence gene expression in response to environmental stimuli. While several post-translational modifications contribute to chromatin accessibility and transcriptional programs, our understanding of the role that specific phosphorylation sites play is limited. In cancer, kinases and phosphatases are commonly deregulated resulting in increased oncogenic signaling and loss of epigenetic regulation. Aberrant epigenetic states are known to promote cellular plasticity and the development of therapeutic resistance in many cancer types, highlighting the importance of these mechanisms to cancer cell phenotypes. Protein Phosphatase 2A (PP2A) is a heterotrimeric holoenzyme that targets a diverse array of cellular proteins. The composition of the PP2A complex influences its cellular targets and activity. For this reason, PP2A can be tumor suppressive or oncogenic depending on cellular context. Understanding the nuances of PP2A regulation and its effect on epigenetic alterations can lead to new therapeutic avenues that afford more specificity and contribute to the growth of personalized medicine in the oncology field. In this review, we summarize the known PP2A-regulated substrates and potential phosphorylation sites that contribute to cancer cell epigenetics and possible strategies to therapeutically leverage this phosphatase to suppress tumor growth.
Collapse
Affiliation(s)
- Samantha L Tinsley
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | | |
Collapse
|
28
|
Llombart V, Mansour MR. Therapeutic targeting of "undruggable" MYC. EBioMedicine 2022; 75:103756. [PMID: 34942444 PMCID: PMC8713111 DOI: 10.1016/j.ebiom.2021.103756] [Citation(s) in RCA: 257] [Impact Index Per Article: 85.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/23/2021] [Accepted: 12/01/2021] [Indexed: 12/13/2022] Open
Abstract
c-MYC controls global gene expression and regulates cell proliferation, cell differentiation, cell cycle, metabolism and apoptosis. According to some estimates, MYC is dysregulated in ≈70% of human cancers and strong evidence implicates aberrantly expressed MYC in both tumor initiation and maintenance. In vivo studies show that MYC inhibition elicits a prominent anti-proliferative effect and sustained tumor regression while any alteration on healthy tissue remains reversible. This opens an exploitable window for treatment that makes MYC one of the most appealing therapeutic targets for cancer drug development. This review describes the main functional and structural features of the protein structure of MYC and provides a general overview of the most relevant or recently identified interactors that modulate MYC oncogenic activity. This review also summarizes the different approaches aiming to abrogate MYC oncogenic function, with a particular focus on the prototype inhibitors designed for the direct and indirect targeting of MYC.
Collapse
Affiliation(s)
- Victor Llombart
- UCL Cancer Institute, University College London, Department of Haematology, London WC1E 6DD, UK
| | - Marc R Mansour
- UCL Cancer Institute, University College London, Department of Haematology, London WC1E 6DD, UK; UCL Great Ormond Street Institute of Child Health, Developmental Biology and Cancer, London, UK.
| |
Collapse
|
29
|
Sergienko NM, Donner DG, Delbridge LMD, McMullen JR, Weeks KL. Protein phosphatase 2A in the healthy and failing heart: New insights and therapeutic opportunities. Cell Signal 2021; 91:110213. [PMID: 34902541 DOI: 10.1016/j.cellsig.2021.110213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 02/06/2023]
Abstract
Protein phosphatases have emerged as critical regulators of phosphoprotein homeostasis in settings of health and disease. Protein phosphatase 2A (PP2A) encompasses a large subfamily of enzymes that remove phosphate groups from serine/threonine residues within phosphoproteins. The heterogeneity in PP2A structure, which arises from the grouping of different catalytic, scaffolding and regulatory subunit isoforms, creates distinct populations of catalytically active enzymes (i.e. holoenzymes) that localise to different parts of the cell. This structural complexity, combined with other regulatory mechanisms, such as interaction of PP2A heterotrimers with accessory proteins and post-translational modification of the catalytic and/or regulatory subunits, enables PP2A holoenzymes to target phosphoprotein substrates in a highly specific manner. In this review, we summarise the roles of PP2A in cardiac physiology and disease. PP2A modulates numerous processes that are vital for heart function including calcium handling, contractility, β-adrenergic signalling, metabolism and transcription. Dysregulation of PP2A has been observed in human cardiac disease settings, including heart failure and atrial fibrillation. Efforts are underway, particularly in the cancer field, to develop therapeutics targeting PP2A activity. The development of small molecule activators of PP2A (SMAPs) and other compounds that selectively target specific PP2A holoenzymes (e.g. PP2A/B56α and PP2A/B56ε) will improve understanding of the function of different PP2A species in the heart, and may lead to the development of therapeutics for normalising aberrant protein phosphorylation in settings of cardiac remodelling and dysfunction.
Collapse
Affiliation(s)
- Nicola M Sergienko
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Central Clinical School, Monash University, Clayton VIC 3800, Australia
| | - Daniel G Donner
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville VIC 3010, Australia
| | - Lea M D Delbridge
- Department of Anatomy and Physiology, The University of Melbourne, Parkville VIC 3010, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville VIC 3010, Australia; Department of Physiology and Department of Medicine Alfred Hospital, Monash University, Clayton VIC 3800, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora VIC 3086, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton VIC 3800, Australia.
| | - Kate L Weeks
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Department of Anatomy and Physiology, The University of Melbourne, Parkville VIC 3010, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville VIC 3010, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton VIC 3800, Australia.
| |
Collapse
|
30
|
Targeting T-type channels in cancer: What is on and what is off? Drug Discov Today 2021; 27:743-758. [PMID: 34838727 DOI: 10.1016/j.drudis.2021.11.021] [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] [Received: 06/17/2021] [Revised: 10/10/2021] [Accepted: 11/18/2021] [Indexed: 12/27/2022]
Abstract
Over the past 20 years, various studies have demonstrated a pivotal role of T-type calcium channels (TTCCs) in tumor progression. Cytotoxic effects of TTCC pharmacological blockers have been reported in vitro and in preclinical models. However, their roles in cancer physiology are only beginning to be understood. In this review, we discuss evidence for the signaling pathways and cellular processes stemming from TTCC activity, mainly inferred by inverse reasoning from pharmacological blocks and, only in a few studies, by gene silencing or channel activation. A thorough analysis indicates that drug-induced cytotoxicity is partially an off-target effect. Dissection of on/off-target activity is paramount to elucidate the physiological roles of TTCCs, and to deliver efficacious therapies suited to different cancer types and stages.
Collapse
|
31
|
Allosteric activation of PP2A inhibits experimental abdominal aortic aneurysm. Clin Sci (Lond) 2021; 135:2085-2097. [PMID: 34402501 DOI: 10.1042/cs20210315] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 08/06/2021] [Accepted: 08/17/2021] [Indexed: 12/22/2022]
Abstract
Although extremely important, the molecular mechanisms that govern aortic aneurysm (AA) formation and progression are still poorly understood. This deficit represents a critical roadblock toward the development of effective pharmaceutical therapies for the treatment of AA. While dysregulation of protein phosphatase 2A (PP2A) is thought to play a role in cardiovascular disease, its role in aortic aneurysm is unknown. The objective of the present study is to test the hypothesis that PP2A regulates abdominal aortic aneurysm (AAA) progression in a murine model. In an angiotensin II-induced AAA murine model, the PP2A inhibitor, LB-100, markedly accelerated AAA progression as demonstrated by increased abdominal aortic dilation and mortality. AAA progression was associated with elevated inflammation and extracellular matrix fragmentation, concomitant with increases in both metalloproteinase activity and reactive oxygen species production. Conversely, administration of a novel class of small molecule activators of PP2A (SMAPs) resulted in an antithetical effect. SMAPs effectively reduced AAA incidence along with the corresponding pathologies that were increased with LB-100 treatment. Mechanistically, modulation of PP2A activities in vivo functioned in part via alteration of the ERK1/2 and NFκB signaling pathways, known regulators of AAA progression. These studies, for the first time, demonstrate a role of PP2A in AAA etiology and demonstrate that PP2A activation may represent a novel strategy for the treatment of abdominal aortic aneurysms.
Collapse
|
32
|
Elevating CDCA3 Levels Enhances Tyrosine Kinase Inhibitor Sensitivity in TKI-Resistant EGFR Mutant Non-Small-Cell Lung Cancer. Cancers (Basel) 2021; 13:cancers13184651. [PMID: 34572879 PMCID: PMC8466783 DOI: 10.3390/cancers13184651] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/25/2021] [Accepted: 08/31/2021] [Indexed: 12/24/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are the first-line therapy for non-small-cell lung cancers (NSCLC) that harbour sensitising mutations within the epidermal growth factor receptor (EGFR). However, resistance remains a key issue, with tumour relapse likely to occur. We have previously identified that cell division cycle-associated protein 3 (CDCA3) is elevated in adenocarcinoma (LUAD) and correlates with sensitivity to platinum-based chemotherapy. Herein, we explored whether CDCA3 levels were associated with EGFR mutant LUAD and TKI response. We demonstrate that in a small-cohort tissue microarray and in vitro LUAD cell line panel, CDCA3 protein levels are elevated in EGFR mutant NSCLC as a result of increased protein stability downstream of receptor tyrosine kinase signalling. Here, CDCA3 protein levels correlated with TKI potency, whereby CDCA3high EGFR mutant NSCLC cells were most sensitive. Consistently, ectopic overexpression or inhibition of casein kinase 2 using CX-4945, which pharmacologically prevents CDCA3 degradation, upregulated CDCA3 levels and the response of T790M(+) H1975 cells and two models of acquired resistance to TKIs. Accordingly, it is possible that strategies to upregulate CDCA3 levels, particularly in CDCA3low tumours or upon the emergence of therapy resistance, might improve the response to EGFR TKIs and benefit patients.
Collapse
|
33
|
Jayashankar V, Selwan E, Hancock SE, Verlande A, Goodson MO, Eckenstein KH, Milinkeviciute G, Hoover BM, Chen B, Fleischman AG, Cramer KS, Hanessian S, Masri S, Turner N, Edinger AL. Drug-like sphingolipid SH-BC-893 opposes ceramide-induced mitochondrial fission and corrects diet-induced obesity. EMBO Mol Med 2021; 13:e13086. [PMID: 34231322 PMCID: PMC8350895 DOI: 10.15252/emmm.202013086] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 06/02/2021] [Accepted: 06/09/2021] [Indexed: 02/06/2023] Open
Abstract
Ceramide-induced mitochondrial fission drives high-fat diet (HFD)-induced obesity. However, molecules targeting mitochondrial dynamics have shown limited benefits in murine obesity models. Here, we reveal that these compounds are either unable to block ceramide-induced mitochondrial fission or require extended incubation periods to be effective. In contrast, targeting endolysosomal trafficking events important for mitochondrial fission rapidly and robustly prevented ceramide-induced disruptions in mitochondrial form and function. By simultaneously inhibiting ARF6- and PIKfyve-dependent trafficking events, the synthetic sphingolipid SH-BC-893 blocked palmitate- and ceramide-induced mitochondrial fission, preserved mitochondrial function, and prevented ER stress in vitro. Similar benefits were observed in the tissues of HFD-fed mice. Within 4 h of oral administration, SH-BC-893 normalized mitochondrial morphology in the livers and brains of HFD-fed mice, improved mitochondrial function in white adipose tissue, and corrected aberrant plasma leptin and adiponectin levels. As an interventional agent, SH-BC-893 restored normal body weight, glucose disposal, and hepatic lipid levels in mice consuming a HFD. In sum, the sphingolipid analog SH-BC-893 robustly and acutely blocks ceramide-induced mitochondrial dysfunction, correcting diet-induced obesity and its metabolic sequelae.
Collapse
Affiliation(s)
- Vaishali Jayashankar
- Department of Developmental and Cell BiologyUniversity of California IrvineIrvineCAUSA
| | - Elizabeth Selwan
- Department of Developmental and Cell BiologyUniversity of California IrvineIrvineCAUSA
| | - Sarah E Hancock
- School of Medical SciencesUniversity of New South WalesSydneyNSWAustralia
| | - Amandine Verlande
- Department of Biological ChemistryUniversity of California IrvineIrvineCAUSA
| | - Maggie O Goodson
- Department of Biological ChemistryUniversity of California IrvineIrvineCAUSA
| | - Kazumi H Eckenstein
- Department of Developmental and Cell BiologyUniversity of California IrvineIrvineCAUSA
| | | | - Brianna M Hoover
- Division of Hematology/OncologyDepartment of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Bin Chen
- Department of ChemistryUniversité de MontréalMontréalQCCanada
| | - Angela G Fleischman
- Division of Hematology/OncologyDepartment of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Karina S Cramer
- Department of Neurobiology and BehaviorUniversity of California IrvineIrvineCAUSA
| | | | - Selma Masri
- Department of Biological ChemistryUniversity of California IrvineIrvineCAUSA
| | - Nigel Turner
- School of Medical SciencesUniversity of New South WalesSydneyNSWAustralia
| | - Aimee L Edinger
- Department of Developmental and Cell BiologyUniversity of California IrvineIrvineCAUSA
| |
Collapse
|
34
|
Targeting protein phosphatase PP2A for cancer therapy: development of allosteric pharmaceutical agents. Clin Sci (Lond) 2021; 135:1545-1556. [PMID: 34192314 DOI: 10.1042/cs20201367] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/15/2021] [Accepted: 07/16/2021] [Indexed: 01/26/2023]
Abstract
Tumor initiation is driven by oncogenes that activate signaling networks for cell proliferation and survival involving protein phosphorylation. Protein kinases in these pathways have proven to be effective targets for pharmaceutical inhibitors that have progressed to the clinic to treat various cancers. Here, we offer a narrative about the development of small molecule modulators of the protein Ser/Thr phosphatase 2A (PP2A) to reduce the activation of cell proliferation and survival pathways. These novel drugs promote the assembly of select heterotrimeric forms of PP2A that act to limit cell proliferation. We discuss the potential for the near-term translation of this approach to the clinic for cancer and other human diseases.
Collapse
|
35
|
Scarpa M, Singh P, Bailey CM, Lee JK, Kapoor S, Lapidus RG, Niyongere S, Sangodkar J, Wang Y, Perrotti D, Narla G, Baer MR. PP2A-activating Drugs Enhance FLT3 Inhibitor Efficacy through AKT Inhibition-Dependent GSK-3β-Mediated c-Myc and Pim-1 Proteasomal Degradation. Mol Cancer Ther 2021; 20:676-690. [PMID: 33568357 PMCID: PMC8027945 DOI: 10.1158/1535-7163.mct-20-0663] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/26/2020] [Indexed: 11/16/2022]
Abstract
Fms-like tyrosine-like kinase 3 internal tandem duplication (FLT3-ITD) is present in acute myeloid leukemia (AML) in 30% of patients and is associated with short disease-free survival. FLT3 inhibitor efficacy is limited and transient but may be enhanced by multitargeting of FLT3-ITD signaling pathways. FLT3-ITD drives both STAT5-dependent transcription of oncogenic Pim-1 kinase and inactivation of the tumor-suppressor protein phosphatase 2A (PP2A), and FLT3-ITD, Pim-1, and PP2A all regulate the c-Myc oncogene. We studied mechanisms of action of cotreatment of FLT3-ITD-expressing cells with FLT3 inhibitors and PP2A-activating drugs (PADs), which are in development. PADs, including FTY720 and DT-061, enhanced FLT3 inhibitor growth suppression and apoptosis induction in FLT3-ITD-expressing cell lines and primary AML cells in vitro and MV4-11 growth suppression in vivo PAD and FLT3 inhibitor cotreatment independently downregulated c-Myc and Pim-1 protein through enhanced proteasomal degradation. c-Myc and Pim-1 downregulation was preceded by AKT inactivation, did not occur in cells expressing myristoylated (constitutively active) AKT1, and could be induced by AKT inhibition. AKT inactivation resulted in activation of GSK-3β, and GSK-3β inhibition blocked downregulation of both c-Myc and Pim-1 by PAD and FLT3 inhibitor cotreatment. GSK-3β activation increased c-Myc proteasomal degradation through c-Myc phosphorylation on T58; infection with c-Myc with T58A substitution, preventing phosphorylation, blocked downregulation of c-Myc by PAD and FLT3 inhibitor cotreatment. GSK-3β also phosphorylated Pim-1L/Pim-1S on S95/S4. Thus, PADs enhance efficacy of FLT3 inhibitors in FLT3-ITD-expressing cells through a novel mechanism involving AKT inhibition-dependent GSK-3β-mediated increased c-Myc and Pim-1 proteasomal degradation.
Collapse
Affiliation(s)
- Mario Scarpa
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center
- Department of Medicine
| | - Prerna Singh
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center
| | - Christopher M Bailey
- Department of Surgery and
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jonelle K Lee
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center
| | - Shivani Kapoor
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center
| | - Rena G Lapidus
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center
- Department of Medicine
| | - Sandrine Niyongere
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center
- Department of Medicine
| | - Jaya Sangodkar
- Division of Genetic Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan
| | - Yin Wang
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center
- Department of Surgery and
- Division of Immunotherapy, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Danilo Perrotti
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center
- Department of Medicine
| | - Goutham Narla
- Division of Genetic Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan
| | - Maria R Baer
- The University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center,
- Department of Medicine
- Veterans Affairs Medical Center, Baltimore, Maryland
| |
Collapse
|
36
|
Tsuji S, Kohyanagi N, Mizuno T, Ohama T, Sato K. Perphenazine exerts antitumor effects on HUT78 cells through Akt dephosphorylation by protein phosphatase 2A. Oncol Lett 2020; 21:113. [PMID: 33376545 PMCID: PMC7751355 DOI: 10.3892/ol.2020.12374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/09/2020] [Indexed: 12/22/2022] Open
Abstract
Sezary syndrome is a rare type of non-Hodgkin lymphoma. Protein phosphatase 2A (PP2A) is an important tumor suppressor whose activity is widely inhibited in a variety of tumors. Recently, reactivation of PP2A has attracted increasing attention as a promising approach for cancer therapy. Phenothiazine anti-psychotic perphenazine (PPZ) exerts antitumor effects by reactivating PP2A. The present study investigated the molecular mechanism underling the antitumor effects of PPZ in the neuroblastoma rat sarcoma oncogene (NRAS)-mutated Sezary syndrome cell line, HUT78. The results of the present study demonstrated that PPZ induced the dephosphorylation of Akt and ERK1/2, and triggered apoptosis in HUT78 cells. In addition, a PP2A inhibitor blocked the PPZ-mediated dephosphorylation of Akt but did not affect that of ERK1/2. The pharmacological inhibition of Akt and ERK1/2 signaling revealed that Akt activity serves an important role in the survival of HUT78 cells. The present data suggested that suppressing Akt activity by PP2A activation may be an attractive antitumor strategy for NRAS-mutated Sezary syndrome.
Collapse
Affiliation(s)
- Shunya Tsuji
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8515, Japan
| | - Naoki Kohyanagi
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8515, Japan
| | - Takuya Mizuno
- The Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8515, Japan
| | - Takashi Ohama
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8515, Japan
| | - Koichi Sato
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8515, Japan
| |
Collapse
|
37
|
Tiwari A, Tashiro K, Dixit A, Soni A, Vogel K, Hall B, Shafqat I, Slaughter J, Param N, Le A, Saunders E, Paithane U, Garcia G, Campos AR, Zettervall J, Carlson M, Starr TK, Marahrens Y, Deshpande AJ, Commisso C, Provenzano PP, Bagchi A. Loss of HIF1A From Pancreatic Cancer Cells Increases Expression of PPP1R1B and Degradation of p53 to Promote Invasion and Metastasis. Gastroenterology 2020; 159:1882-1897.e5. [PMID: 32768595 PMCID: PMC7680408 DOI: 10.1053/j.gastro.2020.07.046] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 07/11/2020] [Accepted: 07/27/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Pancreatic ductal adenocarcinomas (PDACs) are hypovascular, resulting in the up-regulation of hypoxia inducible factor 1 alpha (HIF1A), which promotes the survival of cells under low-oxygen conditions. We studied the roles of HIF1A in the development of pancreatic tumors in mice. METHODS We performed studies with KrasLSL-G12D/+;Trp53LSL-R172H/+;Pdx1-Cre (KPC) mice, KPC mice with labeled pancreatic epithelial cells (EKPC), and EKPC mice with pancreas-specific depletion of HIF1A. Pancreatic and other tissues were collected and analyzed by histology and immunohistochemistry. Cancer cells were cultured from PDACs from mice and analyzed in cell migration and invasion assays and by immunoblots, real-time polymerase chain reaction, and liquid chromatography-mass spectrometry. We performed studies with the human pancreatic cancer cell lines PATU-8988T, BxPC-3, PANC-1, and MiaPACA-2, which have no or low metastatic activity, and PATU-8988S, AsPC-1, SUIT-2 and Capan-1, which have high metastatic activity. Expression of genes was knocked down in primary cancer cells and pancreatic cancer cell lines by using small hairpin RNAs; cells were injected intravenously into immune-competent and NOD/SCID mice, and lung metastases were quantified. We compared levels of messenger RNAs in pancreatic tumors and normal pancreas in The Cancer Genome Atlas. RESULTS EKPC mice with pancreas-specific deletion of HIF1A developed more advanced pancreatic neoplasias and PDACs with more invasion and metastasis, and had significantly shorter survival times, than EKPC mice. Pancreatic cancer cells from these tumors had higher invasive and metastatic activity in culture than cells from tumors of EKPC mice. HIF1A-knockout pancreatic cancer cells had increased expression of protein phosphatase 1 regulatory inhibitor subunit 1B (PPP1R1B). There was an inverse correlation between levels of HIF1A and PPP1R1B in human PDAC tumors; higher expression of PPP1R1B correlated with shorter survival times of patients. Metastatic human pancreatic cancer cell lines had increased levels of PPP1R1B and lower levels of HIF1A compared with nonmetastatic cancer cell lines; knockdown of PPP1R1B significantly reduced the ability of pancreatic cancer cells to form lung metastases in mice. PPP1R1B promoted degradation of p53 by stabilizing phosphorylation of MDM2 at Ser166. CONCLUSIONS HIF1A can act a tumor suppressor by preventing the expression of PPP1R1B and subsequent degradation of the p53 protein in pancreatic cancer cells. Loss of HIF1A from pancreatic cancer cells increases their invasive and metastatic activity.
Collapse
Affiliation(s)
- Ashutosh Tiwari
- Tumor Initiation and Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California.
| | - Kojiro Tashiro
- Tumor Initiation and Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA,These authors contributed equally
| | - Ajay Dixit
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN,These authors contributed equally
| | - Aditi Soni
- Tumor Initiation and Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Keianna Vogel
- Tumor Initiation and Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Bryan Hall
- Tumor Initiation and Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Iram Shafqat
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | | | - Nesteen Param
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - An Le
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Emily Saunders
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Utkarsha Paithane
- Tumor Initiation and Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Guillermina Garcia
- Histology Core, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | | | - Jon Zettervall
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN
| | - Marjorie Carlson
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN
| | - Timothy K. Starr
- Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN
| | - York Marahrens
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN
| | - Aniruddha J. Deshpande
- Tumor Initiation and Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Cosimo Commisso
- Tumor Initiation and Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Paolo P Provenzano
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN
| | - Anindya Bagchi
- Tumor Initiation and Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California.
| |
Collapse
|
38
|
Yang W, Chen N, Li L, Chen X, Liu X, Zhang Y, Cui J. Favorable Immune Microenvironment in Patients with EGFR and MAPK Co-Mutations. LUNG CANCER (AUCKLAND, N.Z.) 2020; 11:59-71. [PMID: 32982525 PMCID: PMC7490071 DOI: 10.2147/lctt.s262822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/13/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE Although EGFR-mutated patients generally do not benefit from checkpoint inhibitors (ICIs), some patients in the KEYNOTE-001 study consistently benefited from this treatment. This study investigated immune microenvironment characteristics to identify the subgroup of patients that may benefit from ICIs. MATERIALS AND METHODS Using data from The Cancer Genome Atlas Program (TCGA) and Cancer Proteome Atlas, TMB and protein level of PD-L1 were explored in the patients with EGFR mutations and wild-type patients. Different patterns of EGFR mutations (according to EGFR co-mutation with different downstream pathway genesets) were used to group EGFR mutation population. Estimated infiltration analyses were used to explore changes in the immune microenvironment. RESULTS This study analyzed somatic mutation data from 1287 patients from five cohorts (TCGA, Broad, The Tumour Sequencing Project, Memorial Sloan Kettering Cancer Center, Catalogue Of Somatic Mutations In Cancer database). The probability of EGFR mutation was approximately 14.30% (184/1287) and the co-mutation rate was 11.41% (21/184) in patients with EGFR mutations. Glycosaminoglycan-related pathways were significantly upregulated in the EGFR mutant group. EGFR-mutated patients had lower TMB and PD-L1 protein levels than those in wild-type patients. Increase immature DCs infiltration and decreased NK CD56dim, T gamma delta, cytotoxic, and Th2 cell infiltration were the main immune changes in EGFR-mutated patients. Patients with EGFR-MAPK co-mutations had higher levels of TMB and PD-L1 protein expression. Meanwhile, the co-mutated patients had a similar immune microenvironment as that in wild-type patients. CONCLUSION In this study, we defined a subgroup of patients with EGFR-MAPK co-mutations. These co-mutated patients may benefit from ICI treatment.
Collapse
Affiliation(s)
- Wang Yang
- The Cancer Center of the First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| | - Naifei Chen
- The Cancer Center of the First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| | - Lingyu Li
- The Cancer Center of the First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| | - Xiao Chen
- The Cancer Center of the First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| | - Xiangliang Liu
- The Cancer Center of the First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| | - Yongfei Zhang
- The Cancer Center of the First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| | - Jiuwei Cui
- The Cancer Center of the First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| |
Collapse
|
39
|
Wei H, Zhang HL, Wang XC, Xie JZ, An DD, Wan L, Wang JZ, Zeng Y, Shu XJ, Westermarck J, Lu YM, Ohlmeyer M, Liu R. Direct Activation of Protein Phosphatase 2A (PP2A) by Tricyclic Sulfonamides Ameliorates Alzheimer's Disease Pathogenesis in Cell and Animal Models. Neurotherapeutics 2020; 17:1087-1103. [PMID: 32096091 PMCID: PMC7609734 DOI: 10.1007/s13311-020-00841-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease for which there are limited therapeutic strategies. Protein phosphatase 2A (PP2A) activity is decreased in AD brains, which promotes the hyperphosphorylation of Tau and APP, thus participate in the formation of neurofibrillary tangles (NFTs) and β-amyloid (Aβ) overproduction. In this study, the effect of synthetic tricyclic sulfonamide PP2A activators (aka SMAPs) on reducing AD-like pathogenesis was evaluated in AD cell models and AD-like hyperhomocysteinemia (HHcy) rat models. SMAPs effectively increased PP2A activity, and decreased tau phosphorylation and Aβ40/42 levels in AD cell models. In HHcy-AD rat models, cognitive impairments induced by HHcy were rescued by SMAP administration. HHcy-induced tau hyperphosphorylation and Aβ overproduction were ameliorated through increasing PP2A activity on compound treatment. Importantly, SMAP therapy also prevented neuronal cell spine loss and neuronal synapse impairment in the hippocampus of HHcy-AD rats. In summary, our data reveal that pharmacological PP2A reactivation may be a novel therapeutic strategy for AD treatment, and that the tricyclic sulfonamides constitute a novel candidate class of AD therapeutic.
Collapse
Affiliation(s)
- Hui Wei
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui-Liang Zhang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Chuan Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Zhao Xie
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan-Dan An
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Wan
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Zeng
- Clinical Laboratory, The Central Hospital of Wuhan, Wuhan, China
| | - Xi-Ji Shu
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China
| | - Jukka Westermarck
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - You-Ming Lu
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Michael Ohlmeyer
- Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Atux Iskay LLC, Plainsboro, NJ, USA.
| | - Rong Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
40
|
Arribas RL, Bordas A, Domènech Omella J, Cedillo JL, Janssens V, Montiel C, de Los Ríos C. An okadaic acid fragment analogue prevents nicotine-induced resistance to cisplatin by recovering PP2A activity in non-small cell lung cancer cells. Bioorg Chem 2020; 100:103874. [PMID: 32361056 DOI: 10.1016/j.bioorg.2020.103874] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/15/2020] [Accepted: 04/21/2020] [Indexed: 12/21/2022]
Abstract
We herein report the design, synthesis, and functional impact of an okadaic acid (OA) small analogue, ITH12680, which restores the activity of phosphoprotein phosphatase 2A (PP2A), whose deficient activity has been implicated in nicotine-mediated tumor progression and chemoresistance in non-small cell lung cancer (NSCLC). For its design, we paid attention to the structure of the PP2A-OA complex, where the C16-C38 OA fragment confers PP2A affinity and selectivity, but it is not involved in the inhibitory effect. Confirming this hypothesis, PP2A activity was not inhibited by ITH12680. By contrast, the compound partially restored OA-exerted PP2A inhibition in vitro. Moreover, flow cytometry and immunoblotting experiments revealed that ITH12680 reversed nicotine-induced cisplatin resistance in NSCLC cells, as it prevented nicotine-induced reduction of Bax expression and inhibited nicotine-mediated activation of cell survival and proliferation kinases, Akt and ERK1/2. Our findings suggest that the rescue of nicotine-inhibited PP2A activity could diminish the resistance to cisplatin treatment observed in NSCLC patients who continue smoking.
Collapse
Affiliation(s)
- Raquel L Arribas
- Department of Pharmacology and Therapeutic, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo, 4, 28029 Madrid, Spain
| | - Anna Bordas
- Department of Pharmacology and Therapeutic, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo, 4, 28029 Madrid, Spain
| | - Judit Domènech Omella
- Department of Cellular & Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, Herestraat 49, B-3000 Leuven, & LKI (Leuven Cancer Institute), Belgium
| | - Jose Luis Cedillo
- Department of Pharmacology and Therapeutic, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo, 4, 28029 Madrid, Spain
| | - Veerle Janssens
- Department of Cellular & Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, Herestraat 49, B-3000 Leuven, & LKI (Leuven Cancer Institute), Belgium
| | - Carmen Montiel
- Department of Pharmacology and Therapeutic, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo, 4, 28029 Madrid, Spain.
| | - Cristóbal de Los Ríos
- Department of Pharmacology and Therapeutic, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo, 4, 28029 Madrid, Spain; Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, C/ Diego de León, 62, 28006 Madrid, Spain.
| |
Collapse
|
41
|
Leonard D, Huang W, Izadmehr S, O'Connor CM, Wiredja DD, Wang Z, Zaware N, Chen Y, Schlatzer DM, Kiselar J, Vasireddi N, Schüchner S, Perl AL, Galsky MD, Xu W, Brautigan DL, Ogris E, Taylor DJ, Narla G. Selective PP2A Enhancement through Biased Heterotrimer Stabilization. Cell 2020; 181:688-701.e16. [PMID: 32315618 DOI: 10.1016/j.cell.2020.03.038] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 12/04/2019] [Accepted: 03/17/2020] [Indexed: 12/15/2022]
Abstract
Impairment of protein phosphatases, including the family of serine/threonine phosphatases designated PP2A, is essential for the pathogenesis of many diseases, including cancer. The ability of PP2A to dephosphorylate hundreds of proteins is regulated by over 40 specificity-determining regulatory "B" subunits that compete for assembly and activation of heterogeneous PP2A heterotrimers. Here, we reveal how a small molecule, DT-061, specifically stabilizes the B56α-PP2A holoenzyme in a fully assembled, active state to dephosphorylate selective substrates, such as its well-known oncogenic target, c-Myc. Our 3.6 Å structure identifies molecular interactions between DT-061 and all three PP2A subunits that prevent dissociation of the active enzyme and highlight inherent mechanisms of PP2A complex assembly. Thus, our findings provide fundamental insights into PP2A complex assembly and regulation, identify a unique interfacial stabilizing mode of action for therapeutic targeting, and aid in the development of phosphatase-based therapeutics tailored against disease specific phospho-protein targets.
Collapse
Affiliation(s)
- Daniel Leonard
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Wei Huang
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Sudeh Izadmehr
- Division of Hematology and Medical Oncology, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Caitlin M O'Connor
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48105, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Danica D Wiredja
- Department of Nutrition, Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Zhizhi Wang
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Nilesh Zaware
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yinghua Chen
- PEPCC Facility, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Daniela M Schlatzer
- Department of Nutrition, Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Janna Kiselar
- Department of Nutrition, Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Nikhil Vasireddi
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Stefan Schüchner
- Center for Medical Biochemistry, Max Perutz Labs, Medical University of Vienna, Dr. Bohr-Gasse 9/2, Vienna 1030, Austria
| | - Abbey L Perl
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Matthew D Galsky
- Division of Hematology and Medical Oncology, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Wenqing Xu
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - David L Brautigan
- Department of Microbiology, Immunology, and Cancer Biology, Center for Cell Signaling, University of Virginia, Charlottesville, VA 22903, USA
| | - Egon Ogris
- Center for Medical Biochemistry, Max Perutz Labs, Medical University of Vienna, Dr. Bohr-Gasse 9/2, Vienna 1030, Austria
| | - Derek J Taylor
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48105, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
42
|
Liu Z, Yoshimi A, Wang J, Cho H, Chun-Wei Lee S, Ki M, Bitner L, Chu T, Shah H, Liu B, Mato AR, Ruvolo P, Fabbri G, Pasqualucci L, Abdel-Wahab O, Rabadan R. Mutations in the RNA Splicing Factor SF3B1 Promote Tumorigenesis through MYC Stabilization. Cancer Discov 2020; 10:806-821. [PMID: 32188705 DOI: 10.1158/2159-8290.cd-19-1330] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/15/2020] [Accepted: 03/12/2020] [Indexed: 11/16/2022]
Abstract
Although mutations in the gene encoding the RNA splicing factor SF3B1 are frequent in multiple cancers, their functional effects and therapeutic dependencies are poorly understood. Here, we characterize 98 tumors and 12 isogenic cell lines harboring SF3B1 hotspot mutations, identifying hundreds of cryptic 3' splice sites common and specific to different cancer types. Regulatory network analysis revealed that the most common SF3B1 mutation activates MYC via effects conserved across human and mouse cells. SF3B1 mutations promote decay of transcripts encoding the protein phosphatase 2A (PP2A) subunit PPP2R5A, increasing MYC S62 and BCL2 S70 phosphorylation which, in turn, promotes MYC protein stability and impair apoptosis, respectively. Genetic PPP2R5A restoration or pharmacologic PP2A activation impaired SF3B1-mutant tumorigenesis, elucidating a therapeutic approach to aberrant splicing by mutant SF3B1. SIGNIFICANCE: Here, we identify that mutations in SF3B1, the most commonly mutated splicing factor gene across cancers, alter splicing of a specific subunit of the PP2A serine/threonine phosphatase complex to confer post-translational MYC and BCL2 activation, which is therapeutically intervenable using an FDA-approved drug.See related commentary by O'Connor and Narla, p. 765.This article is highlighted in the In This Issue feature, p. 747.
Collapse
Affiliation(s)
- Zhaoqi Liu
- Program for Mathematical Genomics, Columbia University, New York, New York.,Departments of Systems Biology and Biomedical Informatics, Columbia University, New York, New York
| | - Akihide Yoshimi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Jiguang Wang
- Division of Life Science, Department of Chemical and Biological Engineering, Center for Systems Biology and Human Health and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Hana Cho
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Stanley Chun-Wei Lee
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michelle Ki
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lillian Bitner
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Timothy Chu
- Program for Mathematical Genomics, Columbia University, New York, New York.,Departments of Systems Biology and Biomedical Informatics, Columbia University, New York, New York
| | - Harshal Shah
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bo Liu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anthony R Mato
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Peter Ruvolo
- Division of Cancer Medicine, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Giulia Fabbri
- Institute for Cancer Genetics, Columbia University, New York, New York
| | - Laura Pasqualucci
- Institute for Cancer Genetics, Columbia University, New York, New York.,Department of Pathology and Cell Biology, and the Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Raul Rabadan
- Program for Mathematical Genomics, Columbia University, New York, New York. .,Departments of Systems Biology and Biomedical Informatics, Columbia University, New York, New York
| |
Collapse
|
43
|
Two-hybrid screening of FAM13A protein partners in lung epithelial cells. BMC Res Notes 2020; 12:804. [PMID: 31900205 PMCID: PMC6942259 DOI: 10.1186/s13104-019-4840-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 12/02/2019] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES Family with sequence similarity 13 member A (FAM13A) genetic variants have been associated with several chronic respiratory diseases including chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF) and lung cancer. The FAM13A protein includes a RhoGTPase activating protein (RhoGAP) domain known to participate in various cellular mechanisms including cell proliferation. While intensive genomic studies have been performed to reveal its involvement in lung diseases, the biological role of FAM13A protein is still not completely elucidated. RESULTS We therefore performed a two-hybrid screening to identify protein partners of FAM13A using a human lung cancer cDNA library. We identified several protein partners with a high confidence score. Researchers in the field of chronic lung diseases may benefit from this two-hybrid screening data which may reveal new research pathways to decipher.
Collapse
|
44
|
Farrington CC, Yuan E, Mazhar S, Izadmehr S, Hurst L, Allen-Petersen BL, Janghorban M, Chung E, Wolczanski G, Galsky M, Sears R, Sangodkar J, Narla G. Protein phosphatase 2A activation as a therapeutic strategy for managing MYC-driven cancers. J Biol Chem 2020. [DOI: 10.1016/s0021-9258(17)49933-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
45
|
Farrington CC, Yuan E, Mazhar S, Izadmehr S, Hurst L, Allen-Petersen BL, Janghorban M, Chung E, Wolczanski G, Galsky M, Sears R, Sangodkar J, Narla G. Protein phosphatase 2A activation as a therapeutic strategy for managing MYC-driven cancers. J Biol Chem 2019; 295:757-770. [PMID: 31822503 DOI: 10.1074/jbc.ra119.011443] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/04/2019] [Indexed: 12/14/2022] Open
Abstract
The tumor suppressor protein phosphatase 2A (PP2A) is a serine/threonine phosphatase whose activity is inhibited in most human cancers. One of the best-characterized PP2A substrates is MYC proto-oncogene basic helix-loop-helix transcription factor (MYC), whose overexpression is commonly associated with aggressive forms of this disease. PP2A directly dephosphorylates MYC, resulting in its degradation. To explore the therapeutic potential of direct PP2A activation in a diverse set of MYC-driven cancers, here we used biochemical assays, recombinant cell lines, gene expression analyses, and immunohistochemistry to evaluate a series of first-in-class small-molecule activators of PP2A (SMAPs) in Burkitt lymphoma, KRAS-driven non-small cell lung cancer, and triple-negative breast cancer. In all tested models of MYC-driven cancer, the SMAP treatment rapidly and persistently inhibited MYC expression through proteasome-mediated degradation, inhibition of MYC transcriptional activity, decreased cancer cell proliferation, and tumor growth inhibition. Importantly, we generated a series of cell lines expressing PP2A-dependent phosphodegron variants of MYC and demonstrated that the antitumorigenic activity of SMAPs depends on MYC degradation. Collectively, the findings presented here indicate a pharmacologically tractable approach to drive MYC degradation by using SMAPs for the management of a broad range of MYC-driven cancers.
Collapse
Affiliation(s)
| | - Eric Yuan
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106
| | - Sahar Mazhar
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106
| | - Sudeh Izadmehr
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Lauren Hurst
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48105
| | - Brittany L Allen-Petersen
- Department of Molecular and Medical Genetics, Oregon Health and Sciences University, Portland, Oregon 97239
| | - Mahnaz Janghorban
- Department of Molecular and Medical Genetics, Oregon Health and Sciences University, Portland, Oregon 97239
| | - Eric Chung
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106
| | - Grace Wolczanski
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48105
| | - Matthew Galsky
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Rosalie Sears
- Department of Molecular and Medical Genetics, Oregon Health and Sciences University, Portland, Oregon 97239
| | - Jaya Sangodkar
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48105
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48105
| |
Collapse
|
46
|
Nader CP, Cidem A, Verrills NM, Ammit AJ. Protein phosphatase 2A (PP2A): a key phosphatase in the progression of chronic obstructive pulmonary disease (COPD) to lung cancer. Respir Res 2019; 20:222. [PMID: 31623614 PMCID: PMC6798356 DOI: 10.1186/s12931-019-1192-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 09/20/2019] [Indexed: 02/06/2023] Open
Abstract
Lung cancer (LC) has the highest relative risk of development as a comorbidity of chronic obstructive pulmonary disease (COPD). The molecular mechanisms that mediate chronic inflammation and lung function impairment in COPD have been identified in LC. This suggests the two diseases are more linked than once thought. Emerging data in relation to a key phosphatase, protein phosphatase 2A (PP2A), and its regulatory role in inflammatory and tumour suppression in both disease settings suggests that it may be critical in the progression of COPD to LC. In this review, we uncover the importance of the functional and active PP2A holoenzyme in the context of both diseases. We describe PP2A inactivation via direct and indirect means and explore the actions of two key PP2A endogenous inhibitors, cancerous inhibitor of PP2A (CIP2A) and inhibitor 2 of PP2A (SET), and the role they play in COPD and LC. We explain how dysregulation of PP2A in COPD creates a favourable inflammatory micro-environment and promotes the initiation and progression of tumour pathogenesis. Finally, we highlight PP2A as a druggable target in the treatment of COPD and LC and demonstrate the potential of PP2A re-activation as a strategy to halt COPD disease progression to LC. Although further studies are required to elucidate if PP2A activity in COPD is a causal link for LC progression, studies focused on the potential of PP2A reactivating agents to reduce the risk of LC formation in COPD patients will be pivotal in improving clinical outcomes for both COPD and LC patients in the future.
Collapse
Affiliation(s)
- Cassandra P Nader
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
| | - Aylin Cidem
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
| | - Nicole M Verrills
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health & Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Alaina J Ammit
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia.
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
47
|
Perl AL, O'Connor CM, Fa P, Mayca Pozo F, Zhang J, Zhang Y, Narla G. Protein phosphatase 2A controls ongoing DNA replication by binding to and regulating cell division cycle 45 (CDC45). J Biol Chem 2019; 294:17043-17059. [PMID: 31562245 DOI: 10.1074/jbc.ra119.010432] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/20/2019] [Indexed: 11/06/2022] Open
Abstract
Genomic replication is a highly regulated process and represents both a potential benefit and liability to rapidly dividing cells; however, the precise post-translational mechanisms regulating genomic replication are incompletely understood. Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase that regulates a diverse array of cellular processes. Here, utilizing both a gain-of-function chemical biology approach and loss-of-function genetic approaches to modulate PP2A activity, we found that PP2A regulates DNA replication. We demonstrate that increased PP2A activity can interrupt ongoing DNA replication, resulting in a prolonged S phase. The impaired replication resulted in a collapse of replication forks, inducing dsDNA breaks, homologous recombination, and a PP2A-dependent replication stress response. Additionally, we show that during replication, PP2A exists in complex with cell division cycle 45 (CDC45) and that increased PP2A activity caused dissociation of CDC45 and polymerase α from the replisome. Furthermore, we found that individuals harboring mutations in the PP2A Aα gene have a higher fraction of genomic alterations, suggesting that PP2A regulates ongoing replication as a mechanism for maintaining genomic integrity. These results reveal a new function for PP2A in regulating ongoing DNA replication and a potential role for PP2A in the intra-S-phase checkpoint.
Collapse
Affiliation(s)
- Abbey L Perl
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106
| | - Caitlin M O'Connor
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106
| | - Pengyan Fa
- Department of Radiation Oncology, Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, Ohio 43210
| | - Franklin Mayca Pozo
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106
| | - Junran Zhang
- Department of Radiation Oncology, Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, Ohio 43210
| | - Youwei Zhang
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106
| | - Goutham Narla
- Department of Internal Medicine, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106 .,Department of Internal Medicine, Division of Genetic Medicine, University of Michigan, Ann Arbor, Michigan 48105
| |
Collapse
|
48
|
Clark AR, Ohlmeyer M. Protein phosphatase 2A as a therapeutic target in inflammation and neurodegeneration. Pharmacol Ther 2019; 201:181-201. [PMID: 31158394 PMCID: PMC6700395 DOI: 10.1016/j.pharmthera.2019.05.016] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
Abstract
Protein phosphatase 2A (PP2A) is a highly complex heterotrimeric enzyme that catalyzes the selective removal of phosphate groups from protein serine and threonine residues. Emerging evidence suggests that it functions as a tumor suppressor by constraining phosphorylation-dependent signalling pathways that regulate cellular transformation and metastasis. Therefore, PP2A-activating drugs (PADs) are being actively sought and investigated as potential novel anti-cancer treatments. Here we explore the concept that PP2A also constrains inflammatory responses through its inhibitory effects on various signalling pathways, suggesting that PADs may be effective in the treatment of inflammation-mediated pathologies.
Collapse
Affiliation(s)
- Andrew R Clark
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom.
| | | |
Collapse
|
49
|
Remmerie M, Janssens V. PP2A: A Promising Biomarker and Therapeutic Target in Endometrial Cancer. Front Oncol 2019; 9:462. [PMID: 31214504 PMCID: PMC6558005 DOI: 10.3389/fonc.2019.00462] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
Over the last decade, the use of targeted therapies has immensely increased in the treatment of cancer. However, treatment for endometrial carcinomas (ECs) has lagged behind, although potential molecular markers have been identified. This is particularly problematic for the type II ECs, since these aggressive tumors are usually not responsive toward the current standard therapies. Therefore, type II ECs are responsible for most EC-related deaths, indicating the need for new treatment options. Interestingly, molecular analyses of type II ECs have uncovered frequent genetic alterations (up to 40%) in PPP2R1A, encoding the Aα subunit of the tumor suppressive heterotrimeric protein phosphatase type 2A (PP2A). PPP2R1A mutations were also reported in type I ECs and other common gynecologic cancers, albeit at much lower frequencies (0-7%). Nevertheless, PP2A inactivation in the latter cancer types is common via other mechanisms, in particular by increased expression of Cancerous Inhibitor of PP2A (CIP2A) and PP2A Methylesterase-1 (PME-1) proteins. In this review, we discuss the therapeutic potential of direct and indirect PP2A targeting compounds, possibly in combination with other anti-cancer drugs, in EC. Furthermore, we investigate the potential of the PP2A status as a predictive and/or prognostic marker for type I and II ECs.
Collapse
Affiliation(s)
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|