1
|
Ding R, Edwards TC, Goswami P, Wilson DJ, Dreis CD, Ye Y, Geraghty RJ, Chen L. p97 Inhibitors Possessing Antiviral Activity Against SARS-CoV-2 and Low Cytotoxicity. Pharmaceuticals (Basel) 2025; 18:131. [PMID: 39861192 PMCID: PMC11768289 DOI: 10.3390/ph18010131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Background: p97 (also known as valosin-containing protein, VCP) is a member of the AAA+ ATPase family and is intimately associated with protein quality control and homeostasis regulation. Therefore, pharmaceutical inhibition of p97 has been actively pursued as an anticancer strategy. Recently, p97 has emerged as an important pro-viral host factor and p97 inhibitors are being evaluated as potential antiviral agents. Methods: We designed and synthesized novel p97 inhibitors based on the rearrangement of the central fused ring of our previously reported p97 inhibitors. These compounds were tested for inhibition of p97, cytotoxicity, and antiviral activity against SARS-CoV-2. Molecular docking was also performed on selected inhibitors to shed light on their binding modes. Results: Among these new p97 inhibitors, two compounds possess enhanced anti-p97 activity over their parent compounds. More significantly, these two inhibitors exhibit strong antiviral activity against SARS-CoV-2 at doses with no significant cytotoxicity. Molecular docking reveals no major change of the binding mode relative to that of their parent compounds, further supporting our design strategy. Conclusions: These compounds are structurally novel p97 inhibitors that display low toxicity and possess promising antiviral activity against SARS-CoV-2 and potentially other viruses. Further structural exploration is therefore justified and improved analogs will serve as useful tools for studying p97 as a promising host antiviral target.
Collapse
Affiliation(s)
- Rui Ding
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA (P.G.); (C.D.D.)
| | - Tiffany C. Edwards
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA (P.G.); (C.D.D.)
| | - Prithwish Goswami
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA (P.G.); (C.D.D.)
| | - Daniel J. Wilson
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA (P.G.); (C.D.D.)
| | - Christine D. Dreis
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA (P.G.); (C.D.D.)
| | - Yihong Ye
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Robert J. Geraghty
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA (P.G.); (C.D.D.)
| | - Liqiang Chen
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA (P.G.); (C.D.D.)
| |
Collapse
|
2
|
Wang S, Nie J, Jiang H, Li A, Zhong N, Tong W, Yao G, Jiang A, Xie X, Zhong Y, Shu Z, Liu J, Yang F, Liu Z. VCP enhances autophagy-related osteosarcoma progression by recruiting USP2 to inhibit ubiquitination and degradation of FASN. Cell Death Dis 2024; 15:788. [PMID: 39489738 PMCID: PMC11532476 DOI: 10.1038/s41419-024-07168-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 10/09/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024]
Abstract
Osteosarcoma (OS) is a highly aggressive malignant tumor with a high rate of disability and mortality rates, and dysregulated autophagy is a crucial factor in cancer. However, the molecular mechanisms that regulate autophagy in OS remain unclear. This study aimed to explore key molecules that affect autophagy in OS and their regulatory mechanisms. We found that fatty acid synthase (FASN) was significantly increased in activated autophagy models of OS and promoted OS proliferation in an autophagy-dependent manner, as detected by LC3 double-labeled fluorescence confocal microscopy, western blotting, transmission electron microscopy (TEM), and cell functional experiments. Furthermore, co-immunoprecipitation combined with mass spectrometry (Co-IP/MS), ubiquitination modification, molecular docking, and protein truncation methods were used to identify FASN-interacting proteins and analyze their effects on OS. Valosin-containing protein (VCP) enhanced the FASN stability by recruiting ubiquitin specific peptidase-2 (USP2) to remove the K48-linked ubiquitin chains from FASN; domain 2 of VCP and the amino acid sequence () of USP2 were critical for their interactions. Gain- and loss-of-function experiments showed that the inhibition of FASN or USP2 attenuated the stimulatory effect of VCP overexpression on autophagy and the malignant phenotypes of OS cells in vitro and in vivo. Notably, micro-CT indicated that VCP induced severe bone destruction in nude mice, which was abrogated by FASN or USP2 downregulation. In summary, VCP recruits USP2 to stabilize FASN by deubiquitylation, thereby activating autophagy and promoting OS progression. The identification of the VCP/USP2/FASN axis, which mediates autophagy regulation, provides important insights into the underlying mechanisms of OS and offers potential diagnostic and therapeutic strategies for patients with OS.
Collapse
Affiliation(s)
- Shijiang Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Jiangbo Nie
- Department of Orthopedic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Haoxin Jiang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Anan Li
- Department of Orthopedic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Nanshan Zhong
- Basic Medical School of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Weilai Tong
- Department of Orthopedic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Geliang Yao
- Department of Orthopedic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Alan Jiang
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, 330006, People's Republic of China
| | - Xinsheng Xie
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, 330006, People's Republic of China
| | - Yanxin Zhong
- Department of Orthopedic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Zhiguo Shu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Jiaming Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, 330006, People's Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Feng Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China.
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, 330006, People's Republic of China.
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China.
- Postdoctoral Innovation Practice Base, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China.
| | - Zhili Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China.
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Diseases, Nanchang, 330006, People's Republic of China.
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China.
| |
Collapse
|
3
|
Wu X, Wu Q, Hou M, Jiang Y, Li M, Jia G, Yang H, Zhang C. Regenerating Chemotherapeutics through Copper-Based Nanomedicine: Disrupting Protein Homeostasis for Enhanced Tumor Therapy. Adv Healthc Mater 2024; 13:e2401954. [PMID: 39039985 DOI: 10.1002/adhm.202401954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/03/2024] [Indexed: 07/24/2024]
Abstract
The bis-(diethyldithiocarbamate)-copper (CuET), the disulfiram (DSF)-Cu complex, has exhibited noteworthy anti-tumor property. However, its efficacy is compromised due to the inadequate oxidative conditions and the limitation of bioavailable copper. Because CuET can inactivate valosin-containing protein (VCP), a bioinformatic pan-cancer analysis of VCP is first conducted in this study to identify CuET as a promising anticancer drug for diverse cancer types. Then, based on the drug action mechanism, a nanocomposite of CuET and copper oxide (CuO) is designed and fabricated utilizing bovine serum albumin (BSA) as the template (denoted as CuET-CuO@BSA, CCB). CCB manifests peroxidase (POD)-mimicking activity to oxidize the tumor endogenous H2O2 to generate reactive oxygen species (ROS), enhancing the chemotherapy effect of CuET. Furthermore, the cupric ions released after enzymatic reaction can regenerate CuET, which markedly perturbs intracellular protein homeostasis and induces apoptosis of tumor cells. Meanwhile, CCB triggers cuproptosis by inducing the aggregation of lipoylated proteins. The multifaceted action of CCB effectively inhibits tumor progression. Therefore, this study presents an innovative CuET therapeutic strategy that creates an oxidative microenvironment in situ and simultaneously self-supply copper source for CuET regeneration through the combination of CuO nanozyme with CuET, which holds promise for application of CuET for effective tumor therapy.
Collapse
Affiliation(s)
- Xubo Wu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Qinghe Wu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Mengfei Hou
- School of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, China
| | - Yifei Jiang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Meng Li
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Guoping Jia
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Huizhen Yang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Chunfu Zhang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| |
Collapse
|
4
|
Yang E, Fan X, Ye H, Sun X, Ji Q, Ding Q, Zhong S, Zhao S, Xuan C, Fang M, Ding X, Cao J. Exploring the role of ubiquitin regulatory X domain family proteins in cancers: bioinformatics insights, mechanisms, and implications for therapy. J Transl Med 2024; 22:157. [PMID: 38365777 PMCID: PMC10870615 DOI: 10.1186/s12967-024-04890-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/13/2024] [Indexed: 02/18/2024] Open
Abstract
UBXD family (UBXDF), a group of proteins containing ubiquitin regulatory X (UBX) domains, play a crucial role in the imbalance of proliferation and apoptotic in cancer. In this study, we summarised bioinformatics proof on multi-omics databases and literature on UBXDF's effects on cancer. Bioinformatics analysis revealed that Fas-associated factor 1 (FAF1) has the largest number of gene alterations in the UBXD family and has been linked to survival and cancer progression in many cancers. UBXDF may affect tumour microenvironment (TME) and drugtherapy and should be investigated in the future. We also summarised the experimental evidence of the mechanism of UBXDF in cancer, both in vitro and in vivo, as well as its application in clinical and targeted drugs. We compared bioinformatics and literature to provide a multi-omics insight into UBXDF in cancers, review proof and mechanism of UBXDF effects on cancers, and prospect future research directions in-depth. We hope that this paper will be helpful for direct cancer-related UBXDF studies.
Collapse
Affiliation(s)
- Enyu Yang
- School of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xiaowei Fan
- School of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Haihan Ye
- School of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xiaoyang Sun
- School of Biological Sciences, The University of Hong Kong, Hong Kong , 999077, Special Administrative Region, China
| | - Qing Ji
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Department of Head and Neck and Rare Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Qianyun Ding
- Department of 'A', The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shulian Zhong
- Zhejiang Sci-Tech University Hospital, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Shuo Zhao
- School of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Cheng Xuan
- School of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Meiyu Fang
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Department of Head and Neck and Rare Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| | - Xianfeng Ding
- School of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| | - Jun Cao
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Department of Head and Neck and Rare Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| |
Collapse
|
5
|
Braxton JR, Southworth DR. Structural insights of the p97/VCP AAA+ ATPase: How adapter interactions coordinate diverse cellular functionality. J Biol Chem 2023; 299:105182. [PMID: 37611827 PMCID: PMC10641518 DOI: 10.1016/j.jbc.2023.105182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/05/2023] [Accepted: 08/14/2023] [Indexed: 08/25/2023] Open
Abstract
p97/valosin-containing protein is an essential eukaryotic AAA+ ATPase with diverse functions including protein homeostasis, membrane remodeling, and chromatin regulation. Dysregulation of p97 function causes severe neurodegenerative disease and is associated with cancer, making this protein a significant therapeutic target. p97 extracts polypeptide substrates from macromolecular assemblies by hydrolysis-driven translocation through its central pore. Growing evidence indicates that this activity is highly coordinated by "adapter" partner proteins, of which more than 30 have been identified and are commonly described to facilitate translocation through substrate recruitment or modification. In so doing, these adapters enable critical p97-dependent functions such as extraction of misfolded proteins from the endoplasmic reticulum or mitochondria, and are likely the reason for the extreme functional diversity of p97 relative to other AAA+ translocases. Here, we review the known functions of adapter proteins and highlight recent structural and biochemical advances that have begun to reveal the diverse molecular bases for adapter-mediated regulation of p97 function. These studies suggest that the range of mechanisms by which p97 activity is controlled is vastly underexplored with significant advances possible for understanding p97 regulation by the most known adapters.
Collapse
Affiliation(s)
- Julian R Braxton
- Graduate Program in Chemistry and Chemical Biology, University of California, San Francisco, San Francisco, California, USA; Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, California, USA
| | - Daniel R Southworth
- Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, California, USA.
| |
Collapse
|
6
|
Huang Y, Wang F, Lin X, Li Q, Lu Y, Zhang J, Shen X, Tan J, Qin Z, Chen J, Chen X, Pan G, Wang X, Zeng Y, Yang S, Liu J, Xing F, Li K, Zhang H. Nuclear VCP drives colorectal cancer progression by promoting fatty acid oxidation. Proc Natl Acad Sci U S A 2023; 120:e2221653120. [PMID: 37788309 PMCID: PMC10576098 DOI: 10.1073/pnas.2221653120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 08/26/2023] [Indexed: 10/05/2023] Open
Abstract
Fatty acid oxidation (FAO) fuels many cancers. However, knowledge of pathways that drive FAO in cancer remains unclear. Here, we revealed that valosin-containing protein (VCP) upregulates FAO to promote colorectal cancer growth. Mechanistically, nuclear VCP binds to histone deacetylase 1 (HDAC1) and facilitates its degradation, thus promoting the transcription of FAO genes, including the rate-limiting enzyme carnitine palmitoyltransferase 1A (CPT1A). FAO is an alternative fuel for cancer cells in environments exhibiting limited glucose availability. We observed that a VCP inhibitor blocked the upregulation of FAO activity and CPT1A expression triggered by metformin in colorectal cancer (CRC) cells. Combined VCP inhibitor and metformin prove more effective than either agent alone in culture and in vivo. Our study illustrates the molecular mechanism underlying the regulation of FAO by nuclear VCP and demonstrates the potential therapeutic utility of VCP inhibitor and metformin combination treatment for colorectal cancer.
Collapse
Affiliation(s)
- Youwei Huang
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou510632, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital Affiliated with Jinan University, Zhuhai519000, China
- Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou510632, China
| | - Fang Wang
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou510632, China
| | - Xi Lin
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou510632, China
| | - Qing Li
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou510632, China
| | - Yuli Lu
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou510632, China
- Department of Public Health, Shantou Center for Disease Control and Prevention, Shantou515000, China
| | - Jiayu Zhang
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou510655, China
| | - Xi Shen
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou510632, China
| | - Jingyi Tan
- Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou510632, China
| | - Zixi Qin
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou510632, China
| | - Jiahong Chen
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou510632, China
- Department of Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing100191, China
| | - Xueqin Chen
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou510632, China
| | - Guopeng Pan
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou510632, China
| | - Xiangyu Wang
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou510632, China
| | - Yuequan Zeng
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou510632, China
| | - Shangqi Yang
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou510632, China
| | - Jun Liu
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou510632, China
| | - Fan Xing
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou510080, China
| | - Kai Li
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou510655, China
| | - Haipeng Zhang
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou510632, China
| |
Collapse
|
7
|
Shakya A, Liu P, Godek J, McKee NW, Dodson M, Anandhan A, Ooi A, Garcia JGN, Costa M, Chapman E, Zhang DD. The NRF2-p97-NRF2 negative feedback loop. Redox Biol 2023; 65:102839. [PMID: 37573837 PMCID: PMC10428046 DOI: 10.1016/j.redox.2023.102839] [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: 05/19/2023] [Revised: 07/15/2023] [Accepted: 08/04/2023] [Indexed: 08/15/2023] Open
Abstract
p97 is a ubiquitin-targeted ATP-dependent segregase that regulates proteostasis, in addition to a variety of other cellular functions. Previously, we demonstrated that p97 negatively regulates NRF2 by extracting ubiquitylated NRF2 from the KEAP1-CUL3-RBX1 E3 ubiquitin ligase complex, facilitating proteasomal destruction. In the current study, we identified p97 as an NRF2-target gene that contains a functional ARE, indicating the presence of an NRF2-p97-NRF2 negative feedback loop that maintains redox homeostasis. Using CRISPR/Cas9 genome editing, we generated endogenous p97 ARE-mutated BEAS-2B cell lines. These p97 ARE-mutated cell lines exhibit altered expression of p97 and NRF2, as well as a compromised response to NRF2 inducers. Importantly, we also found a positive correlation between NRF2 activation and p97 expression in human cancer patients. Finally, using chronic arsenic-transformed cell lines, we demonstrated a synergistic effect of NRF2 and p97 inhibition in killing cancer cells with high NRF2 and p97 expression. Our study suggests dual upregulation of NRF2 and p97 occurs in certain types of cancers, suggesting that inhibition of both NRF2 and p97 could be a promising treatment strategy for stratified cancer patients.
Collapse
Affiliation(s)
- Aryatara Shakya
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Pengfei Liu
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA; National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jack Godek
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Nicholas W McKee
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Matthew Dodson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Annadurai Anandhan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Aikseng Ooi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Joe G N Garcia
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, FL, 33458, USA
| | - Max Costa
- Departments of Environmental Medicine, and Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10010, USA
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA.
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
8
|
Braxton JR, Altobelli CR, Tucker MR, Tse E, Thwin AC, Arkin MR, Southworth DR. The p97/VCP adapter UBXD1 drives AAA+ remodeling and ring opening through multi-domain tethered interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.15.540864. [PMID: 37292947 PMCID: PMC10245715 DOI: 10.1101/2023.05.15.540864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
p97/VCP is an essential cytosolic AAA+ ATPase hexamer that extracts and unfolds substrate polypeptides during protein homeostasis and degradation. Distinct sets of p97 adapters guide cellular functions but their roles in direct control of the hexamer are unclear. The UBXD1 adapter localizes with p97 in critical mitochondria and lysosome clearance pathways and contains multiple p97-interacting domains. We identify UBXD1 as a potent p97 ATPase inhibitor and report structures of intact p97:UBXD1 complexes that reveal extensive UBXD1 contacts across p97 and an asymmetric remodeling of the hexamer. Conserved VIM, UBX, and PUB domains tether adjacent protomers while a connecting strand forms an N-terminal domain lariat with a helix wedged at the interprotomer interface. An additional VIM-connecting helix binds along the second AAA+ domain. Together these contacts split the hexamer into a ring-open conformation. Structures, mutagenesis, and comparisons to other adapters further reveal how adapters containing conserved p97-remodeling motifs regulate p97 ATPase activity and structure.
Collapse
Affiliation(s)
- Julian R. Braxton
- Graduate Program in Chemistry and Chemical Biology; University of California, San Francisco; San Francisco, CA 94158, USA
- Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases; University of California, San Francisco; San Francisco, CA 94158, USA
| | - Chad R. Altobelli
- Graduate Program in Chemistry and Chemical Biology; University of California, San Francisco; San Francisco, CA 94158, USA
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center; University of California, San Francisco; San Francisco, CA 94158, USA
| | - Maxwell R. Tucker
- Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases; University of California, San Francisco; San Francisco, CA 94158, USA
- Graduate Program in Biophysics; University of California, San Francisco; San Francisco, CA 94158, USA
| | - Eric Tse
- Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases; University of California, San Francisco; San Francisco, CA 94158, USA
| | - Aye C. Thwin
- Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases; University of California, San Francisco; San Francisco, CA 94158, USA
| | - Michelle R. Arkin
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center; University of California, San Francisco; San Francisco, CA 94158, USA
| | - Daniel R. Southworth
- Department of Biochemistry and Biophysics and Institute for Neurodegenerative Diseases; University of California, San Francisco; San Francisco, CA 94158, USA
| |
Collapse
|
9
|
Zhang TM, Liao L, Yang SY, Huang MY, Zhang YL, Deng L, Hu SY, Yang F, Zhang FL, Shao ZM, Li DQ. TOLLIP-mediated autophagic degradation pathway links the VCP-TMEM63A-DERL1 signaling axis to triple-negative breast cancer progression. Autophagy 2023; 19:805-821. [PMID: 35920704 PMCID: PMC9980475 DOI: 10.1080/15548627.2022.2103992] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most challenging breast cancer subtype to treat due to the lack of effective targeted therapies. Transmembrane (TMEM) proteins represent attractive drug targets for cancer therapy, but biological functions of most members of the TMEM family remain unknown. Here, we report for the first time that TMEM63A (transmembrane protein 63A), a poorly characterized TMEM protein with unknown functions in human cancer, functions as a novel oncogene to promote TNBC cell proliferation, migration, and invasion in vitro and xenograft tumor growth and lung metastasis in vivo. Mechanistic investigations revealed that TMEM63A localizes in endoplasmic reticulum (ER) and lysosome membranes, and interacts with VCP (valosin-containing protein) and its cofactor DERL1 (derlin 1). Furthermore, TMEM63A undergoes autophagy receptor TOLLIP-mediated autophagic degradation and is stabilized by VCP through blocking its lysosomal degradation. Strikingly, TMEM63A in turn stabilizes oncoprotein DERL1 through preventing TOLLIP-mediated autophagic degradation. Notably, pharmacological inhibition of VCP by CB-5083 or knockdown of DERL1 partially abolishes the oncogenic effects of TMEM63A on TNBC progression both in vitro and in vivo. Collectively, these findings uncover a previously unknown functional and mechanistic role for TMEM63A in TNBC progression and provide a new clue for targeting TMEM63A-driven TNBC tumors by using a VCP inhibitor.Abbreviations: ATG16L1, autophagy related 16 like 1; ATG5, autophagy related 5; ATP5F1B/ATP5B, ATP synthase F1 subunit beta; Baf-A1, bafilomycin A1; CALCOCO2/NDP52, calcium binding and coiled-coil domain 2; CANX, calnexin; DERL1, derlin 1; EGFR, epidermal growth factor receptor; ER, endoplasmic reticulum; ERAD, endoplasmic reticulum-associated degradation; HSPA8, heat shock protein family A (Hsp70) member 8; IP, immunoprecipitation; LAMP2A, lysosomal associated membrane protein 2; NBR1, NBR1 autophagy cargo receptor; OPTN, optineurin; RT-qPCR, reverse transcription-quantitative PCR; SQSTM1/p62, sequestosome 1; TAX1BP1, Tax1 binding protein 1; TMEM63A, transmembrane protein 63A; TNBC, triple-negative breast cancer; TOLLIP, toll interacting protein; VCP, valosin containing protein.
Collapse
Affiliation(s)
- Tai-Mei Zhang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai Yangpu, China
| | - Li Liao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai Yangpu, China.,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China
| | - Shao-Ying Yang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai Yangpu, China
| | - Min-Ying Huang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai Yangpu, China
| | - Yin-Ling Zhang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai Yangpu, China.,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China
| | - Ling Deng
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai Yangpu, China
| | - Shu-Yuan Hu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai Yangpu, China
| | - Fan Yang
- Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China
| | - Fang-Lin Zhang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai Yangpu, China.,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China
| | - Zhi-Min Shao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai Yangpu, China.,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China.,Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China.,Shanghai Key Laboratory of Breast Cancer, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China
| | - Da-Qiang Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai Yangpu, China.,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China.,Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China.,Shanghai Key Laboratory of Breast Cancer, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai Medical College, Fudan University, Shanghai, Yangpu, China
| |
Collapse
|
10
|
GLI1, a novel target of the ER stress regulator p97/VCP, promotes ATF6f-mediated activation of XBP1. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194924. [PMID: 36842643 DOI: 10.1016/j.bbagrm.2023.194924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/31/2023] [Accepted: 02/19/2023] [Indexed: 02/28/2023]
Abstract
Upon accumulation of improperly folded proteins in the Endoplasmic Reticulum (ER), the Unfolded Protein Response (UPR) is triggered to restore ER homeostasis. The induction of stress genes is a sine qua non condition for effective adaptive UPR. Although this requirement has been extensively described, the mechanisms underlying this process remain in part uncharacterized. Here, we show that p97/VCP, an AAA+ ATPase known to contribute to ER stress-induced gene expression, regulates the transcription factor GLI1, a primary effector of Hedgehog (Hh) signaling. Under basal (non-ER stress) conditions, GLI1 is repressed by a p97/VCP-HDAC1 complex while upon ER stress GLI1 is induced through a mechanism requiring both USF2 binding and increase histone acetylation at its promoter. Interestingly, the induction of GLI1 was independent of ligand-regulated Hh signaling. Further analysis showed that GLI1 cooperates with ATF6f to induce promoter activity and expression of XBP1, a key transcription factor driving UPR. Overall, our work demonstrates a novel role for GLI1 in the regulation of ER stress gene expression and defines the interplay between p97/VCP, HDAC1 and USF2 as essential players in this process.
Collapse
|
11
|
Wang K, Chen L, Dai X, Ye Z, Zhou C, Zhang CJ, Feng Z. Synthesis and structure-activity relationships of N - (3 - (1H-imidazol-2-yl) phenyl) - 3-phenylpropionamide derivatives as a novel class of covalent inhibitors of p97/VCP ATPase. Eur J Med Chem 2023; 248:115094. [PMID: 36634454 DOI: 10.1016/j.ejmech.2023.115094] [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: 11/10/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/09/2023]
Abstract
Noncovalent inhibitors of p97 have entered clinical studies. Compared with noncovalent inhibitors, covalent inhibitors have unique advantages in maintaining inhibitory effect and improving the resistance of the target. We previously employed the activity-based protein profiling to definitely identify p97 as the protein target of FL-18 that has a unique scaffold of benpropargylamide coupled with an imidazole. In this study, we report a thorough structure-activity-relationship study involving the new scaffold. A total of three rounds of optimization led to the discovery of the most potent covalent inhibitor of p97 to date. A chemical proteomics study indicated that the newly-synthesized compounds still targeted the C522 residue of p97 and retained selectivity among the complicated whole proteome. This study provides a suite of new covalent inhibitors of p97 to assist in its biological study and drug discovery.
Collapse
Affiliation(s)
- Ke Wang
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Xiannongtan Street, Beijing, 100050, PR China
| | - Lianguo Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, PR China
| | - Xinyan Dai
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Xiannongtan Street, Beijing, 100050, PR China
| | - Zi Ye
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Xiannongtan Street, Beijing, 100050, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, PR China
| | - Chuan Zhou
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Xiannongtan Street, Beijing, 100050, PR China
| | - Chong-Jing Zhang
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Xiannongtan Street, Beijing, 100050, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, PR China.
| | - Zhiqiang Feng
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Xiannongtan Street, Beijing, 100050, PR China.
| |
Collapse
|
12
|
Shmara A, Perez-Rosendahl M, Murphy K, Kwon A, Smith C, Kimonis V. A clinicopathologic study of malignancy in VCP-associated multisystem proteinopathy. Orphanet J Rare Dis 2022; 17:272. [PMID: 35841038 PMCID: PMC9287862 DOI: 10.1186/s13023-022-02403-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 06/26/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Valosin containing protein (VCP) is an important protein with many vital functions mostly related to the ubiquitin-proteasome system that provides protein quality control. VCP-associated inclusion body myopathy with Paget disease of bone and frontotemporal dementia, also termed VCP disease and multisystem proteinopathy (MSP 1), is an autosomal dominant disorder caused by monoallelic variants in the VCP gene on human chromosome 9. VCP has also been strongly involved in cancer, with over-activity of VCP found in several cancers such as prostate, pancreatic, endometrial, esophageal cancers and osteosarcoma. Since MSP1 is caused by gain of function variants in the VCP gene, we hypothesized our patients would show increased risk for developing malignancies. We describe cases of 3 rare malignancies and 4 common cancers from a retrospective dataset. RESULTS Upon surveying 106 families with confirmed VCP variants, we found a higher rate of rare tumors including malignant peripheral nerve sheath tumor, anaplastic pleomorphic xanthoastrocytoma and thymoma. Some of these subjects developed cancer before displaying other classic VCP disease manifestations. We also present cases of common cancers; however, we did not find an increased rate compared to the general population. This could be related to the early mortality associated with this disease, since most patients die in their 50-60 s due to respiratory failure or cardiomyopathy which is earlier than the age at which most cancers appear. CONCLUSION This is the first study that expands the phenotype of VCP disease to potentially include rare cancers and highlights the importance of further investigation of the role of VCP in cancer development. The results of this study in VCP disease patients suggest that patients may be at an increased risk for rare tumors. A larger study will determine if patients with VCP disease develop cancer at a higher rate than the general population. If that is the case, they should be followed up more frequently and screened for recurrence and metastasis of their cancer.
Collapse
Affiliation(s)
- Alyaa Shmara
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of California-Irvine, Lab and FEDEX: Hewitt Hall, Rm 2038, Health Sciences Rd., Irvine, CA, 92697, USA
| | | | - Kady Murphy
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of California-Irvine, Lab and FEDEX: Hewitt Hall, Rm 2038, Health Sciences Rd., Irvine, CA, 92697, USA
| | - Ashley Kwon
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of California-Irvine, Lab and FEDEX: Hewitt Hall, Rm 2038, Health Sciences Rd., Irvine, CA, 92697, USA
| | - Charles Smith
- Department of Neurology and Sanders-Brown Center On Aging, University of Kentucky, Lexington, KY, USA
| | - Virginia Kimonis
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of California-Irvine, Lab and FEDEX: Hewitt Hall, Rm 2038, Health Sciences Rd., Irvine, CA, 92697, USA.
| |
Collapse
|
13
|
VCP interaction with HMGB1 promotes hepatocellular carcinoma progression by activating the PI3K/AKT/mTOR pathway. J Transl Med 2022; 20:212. [PMID: 35562734 PMCID: PMC9102726 DOI: 10.1186/s12967-022-03416-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/27/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most common pathological type of liver cancer. Valosin-containing protein (VCP) is a member of the AAA-ATPase family associated with multiple molecular functions and involved in tumor metastasis and prognosis. However, the role of VCP in HCC progression is still unclear. METHODS We examined the expression of VCP in HCC using the RNA sequencing and microarray data from public databases and measured it in clinical samples and cell lines by western blot, and immunohistochemistry (IHC). We also evaluated the correlation between VCP and clinical features. The VCP-interacting proteins were identified by co-immunoprecipitation combined with mass spectrometry (CoIP/MS). The underlying molecular mechanisms were investigated using in vitro and in vivo models of HCC. RESULTS We found that VCP expression is significantly increased in tumor tissues and is associated with advanced TNM stages and poorer prognosis in HCC patients. In vitro analyses revealed that VCP overexpression promoted HCC cell proliferation, migration, and invasion via PI3K/AKT/mTOR pathway activation. Conversely, VCP knockdown resulted in the reverse phenotypes. In vivo studies indicated that up-regulated VCP expression accelerated tumor growth in a subcutaneous HCC model. The D1 domain of VCP and A box of HMGB1 were identified as the critical regions for their interaction, and D1 area was required for the tumor-promoting effects induced by VCP expression. VCP enhanced the protein stability of HMGB1 by decreasing its degradation via ubiquitin-proteasome process. Inhibition of HMGB1 markedly attenuated VCP-mediated HCC progression and downstream activation of PI3K/AKT/mTOR signals. CONCLUSION Collectively, these findings demonstrate that VCP is a potential prognostic biomarker in HCC and exhibits oncogenic roles via PI3K/AKT/mTOR pathway activation. HMGB1 played an essential role in VCP-mediated HCC progression, indicating that VCP and HMGB1 are potential therapeutic targets in human HCC.
Collapse
|
14
|
NMS-873 Leads to Dysfunctional Glycometabolism in A p97-Independent Manner in HCT116 Colon Cancer Cells. Pharmaceutics 2022; 14:pharmaceutics14040764. [PMID: 35456598 PMCID: PMC9024726 DOI: 10.3390/pharmaceutics14040764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/24/2022] [Accepted: 03/26/2022] [Indexed: 12/28/2022] Open
Abstract
Adenosine triphosphate (ATP)–competitive p97 inhibitor CB-5339, the successor of CB-5083, is being evaluated in Phase 1 clinical trials for anti-cancer therapy. Different modes-of-action p97 inhibitors such as allosteric inhibitors are useful to overcome drug-induced resistance, one of the major problems of targeted therapy. We previously demonstrated that allosteric p97 inhibitor NMS-873 can overcome CB-5083-induced resistance in HCT116. Here we employed chemical proteomics and drug-induced thermal proteome changes to identify drug targets, in combination with drug-resistant cell lines to dissect on- and off-target effects. We found that NMS-873 but not CB-5083 affected glycometabolism. By establishing NMS-873-resistant HCT116 cell lines and performing both cell-based and proteomic analysis, we confirmed that NMS-873 dysregulates glycometabolism in a p97-independent manner. We then used proteome integral solubility alteration with a temperature-based method (PISA T) to identify NDUFAF5 as one of the potential targets of NMS-873 in the mitochondrial complex I. We also demonstrated that glycolysis inhibitor 2-DG enhanced the anti-proliferative effect of NMS-873. The polypharmacology of NMS-873 can be advantageous for anti-cancer therapy for colon cancer.
Collapse
|
15
|
Conserved L464 in p97 D1-D2 linker is critical for p97 cofactor regulated ATPase activity. Biochem J 2021; 478:3185-3204. [PMID: 34405853 DOI: 10.1042/bcj20210288] [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: 04/27/2021] [Revised: 08/10/2021] [Accepted: 08/18/2021] [Indexed: 11/17/2022]
Abstract
p97 protein is a highly conserved, abundant, functionally diverse, structurally dynamic homohexameric AAA enzyme-containing N, D1, and D2 domains. A truncated p97 protein containing the N and D1 domains and the D1-D2 linker (ND1L) exhibits 79% of wild-type (WT) ATPase activity whereas the ND1 domain alone without the linker only has 2% of WT activity. To investigate the relationship between the D1-D2 linker and the D1 domain, we produced p97 ND1L mutants and demonstrated that this 22-residue linker region is essential for D1 ATPase activity. The conserved amino acid leucine 464 (L464) is critical for regulating D1 and D2 ATPase activity by p97 cofactors p37, p47, and Npl4-Ufd1 (NU). Changing leucine to alanine, proline, or glutamate increased the maximum rate of ATP turnover (kcat) of p47-regulated ATPase activities for these mutants, but not for WT. p37 and p47 increased the kcat of the proline substituted linker, suggesting that they induced linker conformations facilitating ATP hydrolysis. NU inhibited D1 ATPase activities of WT and mutant ND1L proteins, but activated D2 ATPase activity of full-length p97. To further understand the mutant mechanism, we used single-particle cryo-EM to visualize the full-length p97L464P and revealed the conformational change of the D1-D2 linker, resulting in a movement of the helix-turn-helix motif (543-569). Taken together with the biochemical and structural results we conclude that the linker helps maintain D1 in a competent conformation and relays the communication to/from the N-domain to the D1 and D2 ATPase domains, which are ∼50 Å away.
Collapse
|
16
|
p97/VCP is highly expressed in the stem-like cells of breast cancer and controls cancer stemness partly through the unfolded protein response. Cell Death Dis 2021; 12:286. [PMID: 33731668 PMCID: PMC7969628 DOI: 10.1038/s41419-021-03555-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 02/02/2021] [Accepted: 02/15/2021] [Indexed: 02/07/2023]
Abstract
p97/VCP, an evolutionarily concerned ATPase, partakes in multiple cellular proteostatic processes, including the endoplasmic reticulum (ER)-associated protein degradation (ERAD). Elevated expression of p97 is common in many cancers and is often associated with poor survival. Here we report that the levels of p97 positively correlated with the histological grade, tumor size, and lymph node metastasis in breast cancers. We further examined p97 expression in the stem-like cancer cells or cancer stem cells (CSCs), a cell population that purportedly underscores cancer initiation, therapeutic resistance, and recurrence. We found that p97 was consistently at a higher level in the CD44+/CD24-, ALDH+, or PKH26+ CSC populations than the respective non-CSC populations in human breast cancer tissues and cancer cell lines and p97 expression also positively correlated with that of SOX2, another CSC marker. To assess the role of p97 in breast cancers, cancer proliferation, mammosphere, and orthotopic growth were analyzed. Similarly as p97 depletion, two pharmacological inhibitors, which targets the ER-associated p97 or globally inhibits p97's ATPase activity, markedly reduced cancer growth and the CSC population. Importantly, depletion or inhibition of p97 greatly suppressed the proliferation of the ALDH+ CSCs and the CSC-enriched mammospheres, while exhibiting much less or insignificant inhibitory effects on the non-CSC cancer cells. Comparable phenotypes produced by blocking ERAD suggest that ER proteostasis is essential for the CSC integrity. Loss of p97 gravely activated the unfolded protein response (UPR) and modulated the expression of multiple stemness and pluripotency regulators, including C/EBPδ, c-MYC, SOX2, and SKP2, which collectively contributed to the demise of CSCs. In summary, p97 controls the breast CSC integrity through multiple targets, many of which directly affect cancer stemness and are induced by UPR activation. Our findings highlight the importance of p97 and ER proteostasis in CSC biology and anticancer therapy.
Collapse
|
17
|
Zhang G, Li S, Wang F, Jones AC, Goldberg AFG, Lin B, Virgil S, Stoltz BM, Deshaies RJ, Chou TF. A covalent p97/VCP ATPase inhibitor can overcome resistance to CB-5083 and NMS-873 in colorectal cancer cells. Eur J Med Chem 2021; 213:113148. [PMID: 33476933 DOI: 10.1016/j.ejmech.2020.113148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/16/2020] [Accepted: 12/28/2020] [Indexed: 12/18/2022]
Abstract
Small-molecule inhibitors of p97 are useful tools to study p97 function. Human p97 is an important AAA ATPase due to its diverse cellular functions and implication in mediating the turnover of proteins involved in tumorigenesis and virus infections. Multiple p97 inhibitors identified from previous high-throughput screening studies are thiol-reactive compounds targeting Cys522 in the D2 ATP-binding domain. Thus, these findings suggest a potential strategy to develop covalent p97 inhibitors. We first used purified p97 to assay several known covalent kinase inhibitors to determine if they can inhibit ATPase activity. We evaluated their selectivity using our dual reporter cells that can distinguish p97 dependent and independent degradation. We selected a β-nitrostyrene scaffold to further study the structure-activity relationship. In addition, we used p97 structures to design and synthesize analogues of pyrazolo[3,4-d]pyrimidine (PP). We incorporated electrophiles into a PP-like compound 17 (4-amino-1-tert-butyl-3-phenyl pyrazolo[3,4-d]pyrimidine) to generate eight compounds. A selective compound 18 (N-(1-(tert-butyl)-3-phenyl-1H-pyrazolo[3,4-d]pyrimidin-4-yl)acrylamide, PPA) exhibited excellent selectivity in an in vitro ATPase activity assay: IC50 of 0.6 μM, 300 μM, and 100 μM for wild type p97, yeast Cdc48, and N-ethylmaleimide sensitive factor (NSF), respectively. To further examine the importance of Cys522 on the active site pocket during PPA inhibition, C522A and C522T mutants of p97 were purified and shown to increase IC50 values by 100-fold, whereas replacement of Thr532 of yeast Cdc48 with Cysteine decreased the IC50 by 10-fold. The molecular modeling suggested the hydrogen bonds and hydrophobic interactions in addition to the covalent bonding at Cys522 between WT-p97 and PPA. Furthermore, tandem mass spectrometry confirmed formation of a covalent bond between Cys522 and PPA. An anti-proliferation assay indicated that the proliferation of HCT116, HeLa, and RPMI8226 was inhibited by PPA with IC50 of 2.7 μM, 6.1 μM, and 3.4 μM, respectively. In addition, PPA is able to inhibit proliferation of two HCT116 cell lines that are resistant to CB-5083 and NMS-873, respectively. Proteomic analysis of PPA-treated HCT116 revealed Gene Ontology enrichment of known p97 functional pathways such as the protein ubiquitination and the ER to Golgi transport vesicle membrane. In conclusion, we have identified and characterized PPA as a selective covalent p97 inhibitor, which will allow future exploration to improve the potency of p97 inhibitors with different mechanisms of action.
Collapse
Affiliation(s)
- Gang Zhang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, United States
| | - Shan Li
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, United States
| | - Feng Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, United States
| | - Amanda C Jones
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, United States
| | - Alexander F G Goldberg
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, United States
| | - Benjamin Lin
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, United States
| | - Scott Virgil
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, United States
| | - Brian M Stoltz
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, United States.
| | - Raymond J Deshaies
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, United States; Howard Hughes Medical Institute, Chevy Chase, MD, 20815, United States.
| | - Tsui-Fen Chou
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, United States; Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA, 91125, United States.
| |
Collapse
|
18
|
Feng Q, Zheng J, Zhang J, Zhao M. Synthesis and In Vitro Evaluation of 2-[3-(2-Aminoethyl)-1 H-indol-1-yl]- N-benzylquinazolin-4-amine as a Novel p97/VCP Inhibitor Lead Capable of Inducing Apoptosis in Cancer Cells. ACS OMEGA 2020; 5:31784-31791. [PMID: 33344832 PMCID: PMC7745420 DOI: 10.1021/acsomega.0c04478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/17/2020] [Indexed: 06/12/2023]
Abstract
P97/VCP, an endoplasmic reticulum associated protein, belongs to AAA ATPase family, ubiquitous ATPases associated with various cellular activities. Recent research has elucidated the roles of p97/VCP and evaluated its potential as a therapeutic target for some kinds of cancer diseases. We screened the small molecule compounds from a previously established library and found promise in the compound 2-[3-(2-aminoethyl)-1H-indol-1-yl]-N-benzylquinazolin-4-amine (FQ393). Data from docking simulation indicates FQ393 acts as an ATP competitor, and ATPase activity assays showed FQ393 was an inhibitor of p97/VCP. Furthermore, in vitro FQ393 is able to promote apoptosis and prohibit proliferation in a variety of cancer cell lines. Using comparative proteomic profiling of HCT-116 cells, we found significantly different canonical KEGG pathways, which revealed that the protein changes in FQ393 groups were associated with p97/VCP or tumor-related pathways. The present data suggests that FQ393 exerts antitumor activity, at least in part through p97/VCP inhibition.
Collapse
Affiliation(s)
- Qiqi Feng
- School
of Pharmaceutical Sciences, Capital Medical
University, Beijing 100069, People’s Republic
of China
- Area
Major Laboratory of Peptide and Small Molecular Drugs, Engineering
Research Center of Endogenous Prophylactic of Ministry of Education
of China, Capital Medical University, Beijing 100069, People’s Republic of China
| | - Jiaying Zheng
- School
of Pharmaceutical Sciences, Capital Medical
University, Beijing 100069, People’s Republic
of China
- Area
Major Laboratory of Peptide and Small Molecular Drugs, Engineering
Research Center of Endogenous Prophylactic of Ministry of Education
of China, Capital Medical University, Beijing 100069, People’s Republic of China
| | - Jie Zhang
- School
of Pharmaceutical Sciences, Capital Medical
University, Beijing 100069, People’s Republic
of China
- Area
Major Laboratory of Peptide and Small Molecular Drugs, Engineering
Research Center of Endogenous Prophylactic of Ministry of Education
of China, Capital Medical University, Beijing 100069, People’s Republic of China
| | - Ming Zhao
- School
of Pharmaceutical Sciences, Capital Medical
University, Beijing 100069, People’s Republic
of China
- Department
of Biomaterials, Beijing Laboratory of Biomedical Materials and Key
Laboratory of Biomedical Materials of Natural Macromolecules, Beijing University of Chemical Technology, Beijing 100026, People’s Republic of China
- Area
Major Laboratory of Peptide and Small Molecular Drugs, Engineering
Research Center of Endogenous Prophylactic of Ministry of Education
of China, Capital Medical University, Beijing 100069, People’s Republic of China
| |
Collapse
|
19
|
Nguyen KM, Busino L. Targeting the E3 ubiquitin ligases DCAF15 and cereblon for cancer therapy. Semin Cancer Biol 2020; 67:53-60. [DOI: 10.1016/j.semcancer.2020.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/02/2020] [Accepted: 03/06/2020] [Indexed: 12/22/2022]
|
20
|
Aguiar BG, Dumas C, Maaroufi H, Padmanabhan PK, Papadopoulou B. The AAA + ATPase valosin-containing protein (VCP)/p97/Cdc48 interaction network in Leishmania. Sci Rep 2020; 10:13135. [PMID: 32753747 PMCID: PMC7403338 DOI: 10.1038/s41598-020-70010-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
Valosin-containing protein (VCP)/p97/Cdc48 is an AAA + ATPase associated with many ubiquitin-dependent cellular pathways that are central to protein quality control. VCP binds various cofactors, which determine pathway selectivity and substrate processing. Here, we used co-immunoprecipitation and mass spectrometry studies coupled to in silico analyses to identify the Leishmania infantum VCP (LiVCP) interactome and to predict molecular interactions between LiVCP and its major cofactors. Our data support a largely conserved VCP protein network in Leishmania including known but also novel interaction partners. Network proteomics analysis confirmed LiVCP-cofactor interactions and provided novel insights into cofactor-specific partners and the diversity of LiVCP complexes, including the well-characterized VCP-UFD1-NPL4 complex. Gene Ontology analysis coupled with digitonin fractionation and immunofluorescence studies support cofactor subcellular compartmentalization with either cytoplasmic or organellar or vacuolar localization. Furthermore, in silico models based on 3D homology modeling and protein-protein docking indicated that the conserved binding modules of LiVCP cofactors, except for NPL4, interact with specific binding sites in the hexameric LiVCP protein, similarly to their eukaryotic orthologs. Altogether, these results allowed us to build the first VCP protein interaction network in parasitic protozoa through the identification of known and novel interacting partners potentially associated with distinct VCP complexes.
Collapse
Affiliation(s)
- Bruno Guedes Aguiar
- Division of Infectious Disease and Immunity, CHU de Quebec Research Center-Laval University, 2705 Laurier Blvd, Quebec, QC, G1V 4G2, Canada
- Department of Microbiology-Infectious Disease and Immunology, Faculty of Medicine, University Laval, Quebec, QC, G1V 4G2, Canada
- Department of Community Medicine, Federal University of Piauí, Teresina, Brazil
| | - Carole Dumas
- Division of Infectious Disease and Immunity, CHU de Quebec Research Center-Laval University, 2705 Laurier Blvd, Quebec, QC, G1V 4G2, Canada
- Department of Microbiology-Infectious Disease and Immunology, Faculty of Medicine, University Laval, Quebec, QC, G1V 4G2, Canada
| | - Halim Maaroufi
- Institut de Biologie Intégrative Et Des Systèmes (IBIS), Laval University, Quebec, QC, Canada
| | - Prasad K Padmanabhan
- Division of Infectious Disease and Immunity, CHU de Quebec Research Center-Laval University, 2705 Laurier Blvd, Quebec, QC, G1V 4G2, Canada
- Department of Microbiology-Infectious Disease and Immunology, Faculty of Medicine, University Laval, Quebec, QC, G1V 4G2, Canada
| | - Barbara Papadopoulou
- Division of Infectious Disease and Immunity, CHU de Quebec Research Center-Laval University, 2705 Laurier Blvd, Quebec, QC, G1V 4G2, Canada.
- Department of Microbiology-Infectious Disease and Immunology, Faculty of Medicine, University Laval, Quebec, QC, G1V 4G2, Canada.
| |
Collapse
|
21
|
Abstract
p97 belongs to the functional diverse superfamily of AAA+ (ATPases Associated with diverse cellular Activities) ATPases and is characterized by an N-terminal regulatory domain and two stacked hexameric ATPase domains forming a central protein conducting channel. p97 is highly versatile and has key functions in maintaining protein homeostasis including protein quality control mechanisms like the ubiquitin proteasome system (UPS) and autophagy to disassemble polyubiquitylated proteins from chromatin, membranes, macromolecular protein complexes and aggregates which are either degraded by the proteasome or recycled. p97 can use energy derived from ATP hydrolysis to catalyze substrate unfolding and threading through its central channel. The function of p97 in a large variety of different cellular contexts is reflected by its simultaneous association with different cofactors, which are involved in substrate recognition and processing, thus leading to the formation of transient multi-protein complexes. Dysregulation in protein homeostasis and proteotoxic stress are often involved in the development of cancer and neurological diseases and targeting the UPS including p97 in cancer is a well-established pharmacological strategy. In this chapter we will describe structural and functional aspects of the p97 interactome in regulating diverse cellular processes and will discuss the role of p97 in targeted cancer therapy.
Collapse
|
22
|
Khong ZJ, Lai SK, Koh CG, Geifman-Shochat S, Li HY. A novel function of AAA-ATPase p97/VCP in the regulation of cell motility. Oncotarget 2020; 11:74-85. [PMID: 32002125 PMCID: PMC6967774 DOI: 10.18632/oncotarget.27419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/21/2019] [Indexed: 11/25/2022] Open
Abstract
High level of the multifunctional AAA-ATPase p97/VCP is often correlated to the development of cancer; however, the underlying mechanism is not understood completely. Here, we report a novel function of p97/VCP in actin regulation and cell motility. We found that loss of p97/VCP promotes stabilization of F-actin, which cannot be reversed by actin-destabilizing agent, Cytochalasin D. Live-cell imaging demonstrated reduced actin dynamics in p97/VCP-knockdown cells, leading to compromised cell motility. We further examined the underlying mechanism and found elevated RhoA protein levels along with increased phosphorylation of its downstream effectors, ROCK, LIMK, and MLC upon the knockdown of p97/VCP. Since p97/VCP is indispensable in the ubiquitination-dependent protein degradation pathway, we investigated if the loss of p97/VCP hinders the protein degradation of RhoA. Knockdown of p97/VCP resulted in a higher amount of ubiquitinated RhoA, suggesting p97/VCP involvement in the proteasome-dependent protein degradation pathway. Finally, we found that p97/VCP interacts with FBXL19, a molecular chaperone known to guide ubiquitinated RhoA for proteasomal degradation. Reduction of p97/VCP may result in the accumulation of RhoA which, in turn, enhances cytoplasmic F-actin formation. In summary, our study uncovered a novel function of p97/VCP in actin regulation and cell motility via the Rho-ROCK dependent pathway which provides fundamental insights into how p97/VCP is involved in cancer development.
Collapse
Affiliation(s)
- Zi-Jia Khong
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Singapore
| | - Soak-Kuan Lai
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Singapore
| | - Cheng-Gee Koh
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Singapore
| | - Susana Geifman-Shochat
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Singapore
| | - Hoi-Yeung Li
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Singapore
| |
Collapse
|
23
|
Morozov AV, Karpov VL. Proteasomes and Several Aspects of Their Heterogeneity Relevant to Cancer. Front Oncol 2019; 9:761. [PMID: 31456945 PMCID: PMC6700291 DOI: 10.3389/fonc.2019.00761] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/29/2019] [Indexed: 01/19/2023] Open
Abstract
The life of every organism is dependent on the fine-tuned mechanisms of protein synthesis and breakdown. The degradation of most intracellular proteins is performed by the ubiquitin proteasome system (UPS). Proteasomes are central elements of the UPS and represent large multisubunit protein complexes directly responsible for the protein degradation. Accumulating data indicate that there is an intriguing diversity of cellular proteasomes. Different proteasome forms, containing different subunits and attached regulators have been described. In addition, proteasomes specific for a particular tissue were identified. Cancer cells are highly dependent on the proper functioning of the UPS in general, and proteasomes in particular. At the same time, the information regarding the role of different proteasome forms in cancer is limited. This review describes the functional and structural heterogeneity of proteasomes, their association with cancer as well as several established and novel proteasome-directed therapeutic strategies.
Collapse
Affiliation(s)
- Alexey V. Morozov
- Laboratory of Regulation of Intracellular Proteolysis, W.A. Engelhardt Institute of Molecular Biology RAS, Moscow, Russia
| | | |
Collapse
|
24
|
Wu Z, Tantray I, Lim J, Chen S, Li Y, Davis Z, Sitron C, Dong J, Gispert S, Auburger G, Brandman O, Bi X, Snyder M, Lu B. MISTERMINATE Mechanistically Links Mitochondrial Dysfunction with Proteostasis Failure. Mol Cell 2019; 75:835-848.e8. [PMID: 31378462 DOI: 10.1016/j.molcel.2019.06.031] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/04/2019] [Accepted: 06/21/2019] [Indexed: 10/26/2022]
Abstract
Mitochondrial dysfunction and proteostasis failure frequently coexist as hallmarks of neurodegenerative disease. How these pathologies are related is not well understood. Here, we describe a phenomenon termed MISTERMINATE (mitochondrial-stress-induced translational termination impairment and protein carboxyl terminal extension), which mechanistically links mitochondrial dysfunction with proteostasis failure. We show that mitochondrial dysfunction impairs translational termination of nuclear-encoded mitochondrial mRNAs, including complex-I 30kD subunit (C-I30) mRNA, occurring on the mitochondrial surface in Drosophila and mammalian cells. Ribosomes stalled at the normal stop codon continue to add to the C terminus of C-I30 certain amino acids non-coded by mRNA template. C-terminally extended C-I30 is toxic when assembled into C-I and forms aggregates in the cytosol. Enhancing co-translational quality control prevents C-I30 C-terminal extension and rescues mitochondrial and neuromuscular degeneration in a Parkinson's disease model. These findings emphasize the importance of efficient translation termination and reveal unexpected link between mitochondrial health and proteome homeostasis mediated by MISTERMINATE.
Collapse
Affiliation(s)
- Zhihao Wu
- Department of Pathology and Programs in Cancer Biology and Neurosciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Ishaq Tantray
- Department of Pathology and Programs in Cancer Biology and Neurosciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Junghyun Lim
- Department of Pathology and Programs in Cancer Biology and Neurosciences, Stanford University School of Medicine, Stanford, CA, USA; Department of Cancer Biology, Genentech Inc., South San Francisco, CA, USA
| | - Songjie Chen
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Yu Li
- Department of Pathology and Programs in Cancer Biology and Neurosciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Zoe Davis
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Cole Sitron
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Jason Dong
- Department of Pathology and Programs in Cancer Biology and Neurosciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Suzana Gispert
- Experimental Neurology, Goethe University Medical School, Goethe, Germany
| | - Georg Auburger
- Experimental Neurology, Goethe University Medical School, Goethe, Germany
| | - Onn Brandman
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaolin Bi
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Michael Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Bingwei Lu
- Department of Pathology and Programs in Cancer Biology and Neurosciences, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
25
|
Rycenga HB, Wolfe KB, Yeh ES, Long DT. Uncoupling of p97 ATPase activity has a dominant negative effect on protein extraction. Sci Rep 2019; 9:10329. [PMID: 31316150 PMCID: PMC6637110 DOI: 10.1038/s41598-019-46949-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 07/04/2019] [Indexed: 12/13/2022] Open
Abstract
p97 is a highly abundant, homohexameric AAA+ ATPase that performs a variety of essential cellular functions. Characterized as a ubiquitin-selective chaperone, p97 recognizes proteins conjugated to K48-linked polyubiquitin chains and promotes their removal from chromatin and other molecular complexes. Changes in p97 expression or activity are associated with the development of cancer and several related neurodegenerative disorders. Although pathogenic p97 mutations cluster in and around p97’s ATPase domains, mutant proteins display normal or elevated ATPase activity. Here, we show that one of the most common p97 mutations (R155C) retains ATPase activity, but is functionally defective. p97-R155C can be recruited to ubiquitinated substrates on chromatin, but is unable to promote substrate removal. As a result, p97-R155C acts as a dominant negative, blocking protein extraction by a similar mechanism to that observed when p97’s ATPase activity is inhibited or inactivated. However, unlike ATPase-deficient proteins, p97-R155C consumes excess ATP, which can hinder high-energy processes. Together, our results shed new insight into how pathogenic mutations in p97 alter its cellular function, with implications for understanding the etiology and treatment of p97-associated diseases.
Collapse
Affiliation(s)
- Halley B Rycenga
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Kelly B Wolfe
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Elizabeth S Yeh
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - David T Long
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
26
|
Parzych K, Saavedra-García P, Valbuena GN, Al-Sadah HA, Robinson ME, Penfold L, Kuzeva DM, Ruiz-Tellez A, Loaiza S, Holzmann V, Caputo V, Johnson DC, Kaiser MF, Karadimitris A, Lam EWF, Chevet E, Feldhahn N, Keun HC, Auner HW. The coordinated action of VCP/p97 and GCN2 regulates cancer cell metabolism and proteostasis during nutrient limitation. Oncogene 2019; 38:3216-3231. [PMID: 30626938 PMCID: PMC6756015 DOI: 10.1038/s41388-018-0651-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 12/05/2018] [Accepted: 12/07/2018] [Indexed: 12/30/2022]
Abstract
VCP/p97 regulates numerous cellular functions by mediating protein degradation through its segregase activity. Its key role in governing protein homoeostasis has made VCP/p97 an appealing anticancer drug target. Here, we provide evidence that VCP/p97 acts as a regulator of cellular metabolism. We found that VCP/p97 was tied to multiple metabolic processes on the gene expression level in a diverse range of cancer cell lines and in patient-derived multiple myeloma cells. Cellular VCP/p97 dependency to maintain proteostasis was increased under conditions of glucose and glutamine limitation in a range of cancer cell lines from different tissues. Moreover, glutamine depletion led to increased VCP/p97 expression, whereas VCP/p97 inhibition perturbed metabolic processes and intracellular amino acid turnover. GCN2, an amino acid-sensing kinase, attenuated stress signalling and cell death triggered by VCP/p97 inhibition and nutrient shortages and modulated ERK activation, autophagy, and glycolytic metabolite turnover. Together, our data point to an interconnected role of VCP/p97 and GCN2 in maintaining cancer cell metabolic and protein homoeostasis.
Collapse
Affiliation(s)
- Katarzyna Parzych
- Cancer Cell Protein Metabolism Group, Centre for Haematology, Department of Medicine, Imperial College London, London, UK
| | - Paula Saavedra-García
- Cancer Cell Protein Metabolism Group, Centre for Haematology, Department of Medicine, Imperial College London, London, UK
| | - Gabriel N Valbuena
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Hibah A Al-Sadah
- Cancer Cell Protein Metabolism Group, Centre for Haematology, Department of Medicine, Imperial College London, London, UK
| | - Mark E Robinson
- Centre for Haematology, Department of Medicine, Imperial College London, London, UK
| | - Lucy Penfold
- Cancer Cell Protein Metabolism Group, Centre for Haematology, Department of Medicine, Imperial College London, London, UK
| | - Desislava M Kuzeva
- Cancer Cell Protein Metabolism Group, Centre for Haematology, Department of Medicine, Imperial College London, London, UK
| | - Angie Ruiz-Tellez
- Cancer Cell Protein Metabolism Group, Centre for Haematology, Department of Medicine, Imperial College London, London, UK
| | - Sandra Loaiza
- Cancer Cell Protein Metabolism Group, Centre for Haematology, Department of Medicine, Imperial College London, London, UK
| | - Viktoria Holzmann
- Cancer Cell Protein Metabolism Group, Centre for Haematology, Department of Medicine, Imperial College London, London, UK
| | - Valentina Caputo
- Centre for Haematology, Department of Medicine, Imperial College London, London, UK
| | - David C Johnson
- Division of Molecular Pathfology, Institute of Cancer Research, Sutton, UK
| | - Martin F Kaiser
- Division of Molecular Pathfology, Institute of Cancer Research, Sutton, UK
| | | | - Eric W-F Lam
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Eric Chevet
- INSERM U1242, Chemistry, Oncogenesis, Stress, Signaling, Université de Rennes 1, Rennes, France
- Centre de Lutte Contre le Cancer Eugène Marquis Rennes, Rennes, France
| | - Niklas Feldhahn
- Centre for Haematology, Department of Medicine, Imperial College London, London, UK
| | - Hector C Keun
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Holger W Auner
- Cancer Cell Protein Metabolism Group, Centre for Haematology, Department of Medicine, Imperial College London, London, UK.
| |
Collapse
|
27
|
Ding R, Zhang T, Wilson DJ, Xie J, Williams J, Xu Y, Ye Y, Chen L. Discovery of Irreversible p97 Inhibitors. J Med Chem 2019; 62:2814-2829. [PMID: 30830772 DOI: 10.1021/acs.jmedchem.9b00144] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Inhibitors of human p97 (also known as valosin-containing protein) have been actively pursued because of their potential therapeutic applications in cancer and other diseases. However, covalent and irreversible p97 inhibitors have not been well explored. Herein, we report our design, synthesis, and biological evaluation of covalent and irreversible inhibitors of p97. Among an amide and a reverse amide series we synthesized, we have identified a p97 inhibitor whose functional irreversibility has been established both in vitro and in cells. Also importantly, mass spectrometry reveals three potential cysteine residues labeled by this compound, and mutagenesis together with computer modeling suggests Cys522 as a major site, which when modified, could compromise the function of p97. Taken together, this new inhibitor may provide a template for designing more potent p97 inhibitors with covalent and irreversible characteristics.
Collapse
Affiliation(s)
- Rui Ding
- Center for Drug Design, College of Pharmacy , University of Minnesota , Minneapolis , Minnesota 55455 , United States
| | - Ting Zhang
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Daniel J Wilson
- Center for Drug Design, College of Pharmacy , University of Minnesota , Minneapolis , Minnesota 55455 , United States
| | - Jiashu Xie
- Center for Drug Design, College of Pharmacy , University of Minnesota , Minneapolis , Minnesota 55455 , United States
| | - Jessica Williams
- Center for Drug Design, College of Pharmacy , University of Minnesota , Minneapolis , Minnesota 55455 , United States
| | - Yue Xu
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Yihong Ye
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Liqiang Chen
- Center for Drug Design, College of Pharmacy , University of Minnesota , Minneapolis , Minnesota 55455 , United States
| |
Collapse
|
28
|
Ballar Kırmızıbayrak P, Erbaykent Tepedelen B. Endoplazmik retikulum-aracılı protein yıkım yolağında görev yapan p97/VCP ve Ufd1-Npl4 adlı proteinlerin NF-κB yolağına etkisinin prostat kanser hücre hattında incelenmesi. EGE TIP DERGISI 2019. [DOI: 10.19161/etd.418150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
29
|
Guedes Aguiar B, Padmanabhan PK, Dumas C, Papadopoulou B. Valosin-containing protein VCP/p97 is essential for the intracellular development of Leishmania and its survival under heat stress. Cell Microbiol 2018; 20:e12867. [PMID: 29895095 DOI: 10.1111/cmi.12867] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/14/2018] [Accepted: 06/05/2018] [Indexed: 12/15/2022]
Abstract
Valosin-containing protein (VCP)/p97/Cdc48 is one of the best-characterised type II cytosolic AAA+ ATPases most known for their role in ubiquitin-dependent protein quality control. Here, we provide functional insights into the role of the Leishmania VCP/p97 homologue (LiVCP) in the parasite intracellular development. We demonstrate that although LiVCP is an essential gene, Leishmania infantum promastigotes can grow with less VCP. In contrast, growth of axenic and intracellular amastigotes is dramatically affected upon decreased LiVCP levels in heterozygous and temperature sensitive (ts) LiVCP mutants or the expression of dominant negative mutants known to specifically target the second conserved VCP ATPase domain, a major contributor of the VCP overall ATPase activity. Interestingly, these VCP mutants are also unable to survive heat stress, and a ts VCP mutant is defective in amastigote growth. Consistent with LiVCP's essential function in amastigotes, LiVCP messenger ribonucleic acid undergoes 3'Untranslated Region (UTR)-mediated developmental regulation, resulting in higher VCP expression in amastigotes. Furthermore, we show that parasite mutant lines expressing lower VCP levels or dominant negative VCP forms exhibit high accumulation of polyubiquitinated proteins and increased sensitivity to proteotoxic stress, supporting the ubiquitin-selective chaperone function of LiVCP. Together, these results emphasise the crucial role LiVCP plays under heat stress and during the parasite intracellular development.
Collapse
Affiliation(s)
- Bruno Guedes Aguiar
- Research Center in Infectious Diseases, CHU de Quebec Research Center-University Laval, Quebec, Canada.,Department of Microbiology-Infectious Disease and Immunology, Faculty of Medicine, University Laval, Quebec, Canada.,Department of Community Medicine, Federal University of Piauí, Teresina, Brazil
| | - Prasad K Padmanabhan
- Research Center in Infectious Diseases, CHU de Quebec Research Center-University Laval, Quebec, Canada.,Department of Microbiology-Infectious Disease and Immunology, Faculty of Medicine, University Laval, Quebec, Canada
| | - Carole Dumas
- Research Center in Infectious Diseases, CHU de Quebec Research Center-University Laval, Quebec, Canada.,Department of Microbiology-Infectious Disease and Immunology, Faculty of Medicine, University Laval, Quebec, Canada
| | - Barbara Papadopoulou
- Research Center in Infectious Diseases, CHU de Quebec Research Center-University Laval, Quebec, Canada.,Department of Microbiology-Infectious Disease and Immunology, Faculty of Medicine, University Laval, Quebec, Canada
| |
Collapse
|
30
|
Huiting LN, Samaha Y, Zhang GL, Roderick JE, Li B, Anderson NM, Wang YW, Wang L, Laroche F, Choi JW, Liu CT, Kelliher MA, Feng H. UFD1 contributes to MYC-mediated leukemia aggressiveness through suppression of the proapoptotic unfolded protein response. Leukemia 2018; 32:2339-2351. [PMID: 29743725 PMCID: PMC6202254 DOI: 10.1038/s41375-018-0141-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 03/11/2018] [Accepted: 03/15/2018] [Indexed: 02/07/2023]
Abstract
Despite the pivotal role of MYC in tumorigenesis, the mechanisms by which it promotes cancer aggressiveness remain incompletely understood. Here we show that MYC transcriptionally upregulates the ubiquitin fusion degradation 1 (UFD1) gene in T-cell acute lymphoblastic leukemia (T-ALL). Allelic loss of ufd1 in zebrafish induces tumor-cell apoptosis and impairs MYC-driven T-ALL progression but does not affect general health. As the E2 component of an endoplasmic reticulum (ER)-associated degradation (ERAD) complex, UFD1 facilitates the elimination of misfolded/unfolded proteins from the ER. We found that UFD1 inactivation in human T-ALL cells impairs ERAD, exacerbates ER stress, and induces apoptosis. Moreover, we show that UFD1 inactivation promotes the proapoptotic unfolded protein response (UPR) mediated by protein kinase RNA-like ER kinase (PERK). This effect is demonstrated by an upregulation of PERK and its downstream effector C/EBP homologous protein (CHOP), as well as a downregulation of BCL2 and BCLxL. Indeed, CHOP inactivation or BCL2 overexpression is sufficient to rescue tumor-cell apoptosis induced by UFD1 knockdown. Together, our studies identify UFD1 as a critical regulator of the ER stress response and a novel contributor to MYC-mediated leukemia aggressiveness, with implications for targeted therapy in T-ALL and likely other MYC-driven cancers.
Collapse
Affiliation(s)
- L N Huiting
- Departments of Pharmacology and Medicine, Cancer Research Center, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, USA
| | - Y Samaha
- Departments of Pharmacology and Medicine, Cancer Research Center, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, USA
| | - G L Zhang
- Department of Computer Science, Metropolitan College, Boston University, Boston, MA, USA.,Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - J E Roderick
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts School of Medicine, Worcester, MA, USA
| | - B Li
- Departments of Pharmacology and Medicine, Cancer Research Center, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, USA
| | - N M Anderson
- Departments of Pharmacology and Medicine, Cancer Research Center, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, USA
| | - Y W Wang
- Departments of Pharmacology and Medicine, Cancer Research Center, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, USA.,Department of Anatomy and Embryology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei, P. R. China
| | - L Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Fjf Laroche
- Departments of Pharmacology and Medicine, Cancer Research Center, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, USA
| | - J W Choi
- Departments of Pharmacology and Medicine, Cancer Research Center, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, USA
| | - C T Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - M A Kelliher
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts School of Medicine, Worcester, MA, USA
| | - H Feng
- Departments of Pharmacology and Medicine, Cancer Research Center, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
31
|
Heidelberger JB, Voigt A, Borisova ME, Petrosino G, Ruf S, Wagner SA, Beli P. Proteomic profiling of VCP substrates links VCP to K6-linked ubiquitylation and c-Myc function. EMBO Rep 2018; 19:embr.201744754. [PMID: 29467282 DOI: 10.15252/embr.201744754] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 01/17/2018] [Accepted: 01/26/2018] [Indexed: 12/20/2022] Open
Abstract
Valosin-containing protein (VCP) is an evolutionarily conserved ubiquitin-dependent ATPase that mediates the degradation of proteins through the ubiquitin-proteasome pathway. Despite the central role of VCP in the regulation of protein homeostasis, identity and nature of its cellular substrates remain poorly defined. Here, we combined chemical inhibition of VCP and quantitative ubiquitin remnant profiling to assess the effect of VCP inhibition on the ubiquitin-modified proteome and to probe the substrate spectrum of VCP in human cells. We demonstrate that inhibition of VCP perturbs cellular ubiquitylation and increases ubiquitylation of a different subset of proteins compared to proteasome inhibition. VCP inhibition globally upregulates K6-linked ubiquitylation that is dependent on the HECT-type ubiquitin E3 ligase HUWE1. We report ~450 putative VCP substrates, many of which function in nuclear processes, including gene expression, DNA repair and cell cycle. Moreover, we identify that VCP regulates the level and activity of the transcription factor c-Myc.
Collapse
Affiliation(s)
| | - Andrea Voigt
- Institute of Molecular Biology (IMB), Mainz, Germany
| | | | | | - Stefanie Ruf
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Sebastian A Wagner
- Department of Medicine, Hematology/Oncology, Goethe University School of Medicine, Frankfurt, Germany
| | - Petra Beli
- Institute of Molecular Biology (IMB), Mainz, Germany
| |
Collapse
|
32
|
Gareau A, Rico C, Boerboom D, Nadeau ME. In vitro efficacy of a first-generation valosin-containing protein inhibitor (CB-5083) against canine lymphoma. Vet Comp Oncol 2018; 16:311-317. [PMID: 29314493 DOI: 10.1111/vco.12380] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 11/26/2022]
Abstract
Valosin-containing protein (VCP), through its critical role in the maintenance of protein homeostasis, is a promising target for the treatment of several malignancies, including canine lymphoma. CB-5083, a first-in-class VCP inhibitor, exerts cytotoxicity through the induction of irreversible proteotoxic stress and possesses a broad spectrum of anticancer activity. Here, we determined the cytotoxicity CB-5083 in canine lymphoma cells and its mechanism of action in vitro. Canine lymphoma cell lines were treated with varying concentrations of CB-5083 and assessed for viability by trypan blue exclusion and apoptosis by caspase activity assays. The mechanism of CB-5083 action was determined by immunoblotting and RT-qPCR analyses of Lys48 ubiquitination and markers of ER stress (DDIT3), autophagy (SQSTM1, MAP1LC3A) and DNA damage (γH2AX). Unfolded protein response markers were also evaluated by immunoblotting (eIF2α, P-eIF2α) and RT-qPCR (ATF4). CB-5083 treatment resulted in preferential cytotoxicity in canine lymphoma cell lines over control peripheral blood mononuclear cells. CB-5083 rapidly disrupted the ubiquitin-dependent protein degradation system, inducing sustained ER stress as indicated by a dramatic increase in DDIT3. Activation of the unfolded protein response occurred through the increase eIF2α phosphorylation and increased transcription of ATF4, but did not re-establish protein homeostasis. Cells rapidly underwent apoptosis through activation of the caspase cascade. These results further validate VCP as an attractive target for the treatment of canine lymphoma and identify CB-5083 as a novel therapy with clinical potential for this malignancy.
Collapse
Affiliation(s)
- A Gareau
- Faculté de Médecine vétérinaire, Université de Montreal, St-Hyacinthe, Québec, Canada J2C 7C6
| | - C Rico
- Faculté de Médecine vétérinaire, Université de Montreal, St-Hyacinthe, Québec, Canada J2C 7C6
| | - D Boerboom
- Faculté de Médecine vétérinaire, Université de Montreal, St-Hyacinthe, Québec, Canada J2C 7C6
| | - M-E Nadeau
- Faculté de Médecine vétérinaire, Université de Montreal, St-Hyacinthe, Québec, Canada J2C 7C6
| |
Collapse
|
33
|
Creaney J, Dick IM, Leon JS, Robinson BWS. A Proteomic Analysis of the Malignant Mesothelioma Secretome Using iTRAQ. Cancer Genomics Proteomics 2017; 14:103-117. [PMID: 28387650 DOI: 10.21873/cgp.20023] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/28/2017] [Accepted: 02/28/2017] [Indexed: 12/30/2022] Open
Abstract
Backgound/Aim: Malignant mesothelioma (MM) is an aggressive and fatal pleural cancer. The cell secretome offers information allowing insight into the pathogenesis of MM while offering the possibility to identify potential therapeutic targets and biomarkers. In the present study the secretome protein profile of MM cell lines was compared to normal mesothelial cells. MATERIALS AND METHODS Six MM cell lines were compared against three primary mesothelial cell culture preparations using iTRAQ® mass spectrometry. RESULTS MM cell lines more abundantly secreted exosome-associated proteins than mesothelial cells. MM cell secretomes were enriched in proteins that are involved in response to stress, carbon metabolism, biosynthesis of amino acids, antigen processing and presentation and protein processing in the endoplasmic reticulum. CONCLUSION The MM cell secretome is enriched in proteins that are likely to enhance its growth and response to stress and help it inhibit an adaptive immune response. These are potential targets for therapeutic and biomarker discovery.
Collapse
Affiliation(s)
- Jenette Creaney
- National Centre for Asbestos Related Diseases, School of Medicine and Pharmacology, University of Western Australia and Australian Mesothelioma Tissue Bank, Sir Charles Gairdner Hospital, Perth, Western Australia
| | - Ian M Dick
- National Centre for Asbestos Related Diseases, School of Medicine and Pharmacology, University of Western Australia, Sir Charles Gairdner Hospital, Perth, Western Australia
| | - Justine S Leon
- National Centre for Asbestos Related Diseases, School of Medicine and Pharmacology, University of Western Australia, Sir Charles Gairdner Hospital, Perth, Western Australia
| | - Bruce W S Robinson
- National Centre for Asbestos Related Diseases, School of Medicine and Pharmacology, University of Western Australia, Sir Charles Gairdner Hospital, Perth, Western Australia
| |
Collapse
|
34
|
Hänzelmann P, Schindelin H. The Interplay of Cofactor Interactions and Post-translational Modifications in the Regulation of the AAA+ ATPase p97. Front Mol Biosci 2017; 4:21. [PMID: 28451587 PMCID: PMC5389986 DOI: 10.3389/fmolb.2017.00021] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/24/2017] [Indexed: 12/18/2022] Open
Abstract
The hexameric type II AAA ATPase (ATPase associated with various activities) p97 (also referred to as VCP, Cdc48, and Ter94) is critically involved in a variety of cellular activities including pathways such as DNA replication and repair which both involve chromatin remodeling, and is a key player in various protein quality control pathways mediated by the ubiquitin proteasome system as well as autophagy. Correspondingly, p97 has been linked to various pathophysiological states including cancer, neurodegeneration, and premature aging. p97 encompasses an N-terminal domain, two highly conserved ATPase domains and an unstructured C-terminal tail. This enzyme hydrolyzes ATP and utilizes the resulting energy to extract or disassemble protein targets modified with ubiquitin from stable protein assemblies, chromatin and membranes. p97 participates in highly diverse cellular processes and hence its activity is tightly controlled. This is achieved by multiple regulatory cofactors, which either associate with the N-terminal domain or interact with the extreme C-terminus via distinct binding elements and target p97 to specific cellular pathways, sometimes requiring the simultaneous association with more than one cofactor. Most cofactors are recruited to p97 through conserved binding motifs/domains and assist in substrate recognition or processing by providing additional molecular properties. A tight control of p97 cofactor specificity and diversity as well as the assembly of higher-order p97-cofactor complexes is accomplished by various regulatory mechanisms, which include bipartite binding, binding site competition, changes in oligomeric assemblies, and nucleotide-induced conformational changes. Furthermore, post-translational modifications (PTMs) like acetylation, palmitoylation, phosphorylation, SUMOylation, and ubiquitylation of p97 have been reported which further modulate its diverse molecular activities. In this review, we will describe the molecular basis of p97-cofactor specificity/diversity and will discuss how PTMs can modulate p97-cofactor interactions and affect the physiological and patho-physiological functions of p97.
Collapse
Affiliation(s)
- Petra Hänzelmann
- Rudolf Virchow Center for Experimental Biomedicine, University of WürzburgWürzburg, Germany
| | - Hermann Schindelin
- Rudolf Virchow Center for Experimental Biomedicine, University of WürzburgWürzburg, Germany
| |
Collapse
|
35
|
Ding R, Zhang T, Xie J, Williams J, Ye Y, Chen L. Eeyarestatin I derivatives with improved aqueous solubility. Bioorg Med Chem Lett 2016; 26:5177-5181. [DOI: 10.1016/j.bmcl.2016.09.068] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 09/23/2016] [Accepted: 09/28/2016] [Indexed: 12/17/2022]
|
36
|
Walworth K, Bodas M, Campbell RJ, Swanson D, Sharma A, Vij N. Dendrimer-Based Selective Proteostasis-Inhibition Strategy to Control NSCLC Growth and Progression. PLoS One 2016; 11:e0158507. [PMID: 27434122 PMCID: PMC4951140 DOI: 10.1371/journal.pone.0158507] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/16/2016] [Indexed: 02/06/2023] Open
Abstract
Elevated valosin containing protein (VCP/p97) levels promote the progression of non-small cell lung carcinoma (NSCLC). Although many VCP inhibitors are available, most of these therapeutic compounds have low specificity for targeted tumor cell delivery. Hence, the primary aim of this study was to evaluate the in vitro efficacy of dendrimer-encapsulated potent VCP-inhibitor drug in controlling non-small cell lung carcinoma (NSCLC) progression. The VCP inhibitor(s) (either in their pure form or encapsulated in generation-4 PAMAM-dendrimer with hydroxyl surface) were tested for their in vitro efficacy in modulating H1299 (NSCLC cells) proliferation, migration, invasion, apoptosis and cell cycle progression. Our results show that VCP inhibition by DBeQ was significantly more potent than NMS-873 as evident by decreased cell proliferation (p<0.0001, MTT-assay) and migration (p<0.05; scratch-assay), and increased apoptosis (p<0.05; caspase-3/7-assay) as compared to untreated control cells. Next, we found that dendrimer-encapsulated DBeQ (DDNDBeQ) treatment increased ubiquitinated-protein accumulation in soluble protein-fraction (immunoblotting) of H1299 cells as compared to DDN-control, implying the effectiveness of DBeQ in proteostasis-inhibition. We verified by immunostaining that DDNDBeQ treatment increases accumulation of ubiquitinated-proteins that co-localizes with an ER-marker, KDEL. We observed that proteostasis-inhibition with DDNDBeQ, significantly decreased cell migration rate (scratch-assay and transwell-invasion) as compared to the control-DDN treatment (p<0.05). Moreover, DDNDBeQ treatment showed a significant decrease in cell proliferation (p<0.01, MTT-assay) and increased caspase-3/7 mediated apoptotic cell death (p<0.05) as compared to DDN-control. This was further verified by cell cycle analysis (propidium-iodide-staining) that demonstrated significant cell cycle arrest in the G2/M-phase (p<0.001) by DDNDBeQ treatment as compared to control-DDN. Moreover, we confirmed by clonogenic-assay that DDNDBeQ treatment significantly (p<0.001) inhibits H1299 colony-formation as compared to control/DDN. Overall, encapsulation of potent VCP-inhibitor DBeQ into a dendrimer allows selective VCP-mediated proteostasis-inhibition for controlling NSCLC-tumor growth and progression to allow tumor-targeted sustained drug delivery.
Collapse
Affiliation(s)
- Kyla Walworth
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Manish Bodas
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Ryan John Campbell
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Doug Swanson
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Ajit Sharma
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Neeraj Vij
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan, United States of America
- Department of Pediatric Respiratory Sciences, The Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
- * E-mail: ;
| |
Collapse
|
37
|
Wang T, Xu W, Qin M, Yang Y, Bao P, Shen F, Zhang Z, Xu J. Pathogenic Mutations in the Valosin-containing Protein/p97(VCP) N-domain Inhibit the SUMOylation of VCP and Lead to Impaired Stress Response. J Biol Chem 2016; 291:14373-14384. [PMID: 27226613 DOI: 10.1074/jbc.m116.729343] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Indexed: 11/06/2022] Open
Abstract
Valosin-containing protein/p97(VCP) is a hexameric ATPase vital to protein degradation during endoplasmic reticulum stress. It regulates diverse cellular functions including autophagy, chromatin remodeling, and DNA repair. In addition, mutations in VCP cause inclusion body myopathy, Paget disease of the bone, and frontotemporal dementia (IBMPFD), as well as amyotrophic lateral sclerosis. Nevertheless, how the VCP activities were regulated and how the pathogenic mutations affect the function of VCP during stress are not unclear. Here we show that the small ubiquitin-like modifier (SUMO)-ylation of VCP is a normal stress response inhibited by the disease-causing mutations in the N-domain. Under oxidative and endoplasmic reticulum stress conditions, the SUMOylation of VCP facilitates the distribution of VCP to stress granules and nucleus, and promotes the VCP hexamer assembly. In contrast, pathogenic mutations in the VCP N-domain lead to reduced SUMOylation and weakened VCP hexamer formation upon stress. Defective SUMOylation of VCP also causes altered co-factor binding and attenuated endoplasmic reticulum-associated protein degradation. Furthermore, SUMO-defective VCP fails to protect against stress-induced toxicity in Drosophila Therefore, our results have revealed SUMOylation as a molecular signaling switch to regulate the distribution and functions of VCP during stress response, and suggest that deficiency in VCP SUMOylation caused by pathogenic mutations will render cells vulnerable to stress insults.
Collapse
Affiliation(s)
- Tao Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031
| | - Wangchao Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031,; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Meiling Qin
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031
| | - Yi Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031,; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Puhua Bao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031
| | - Fuxiao Shen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031
| | - Zhenlin Zhang
- Department of Osteoporosis and Bone Diseases, Metabolic Bone Disease and Genetic Research Unit, Shanghai Jiao Tong University Affiliated People's No.6 Hospital, Shanghai 200233, China
| | - Jin Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031,.
| |
Collapse
|
38
|
Franz A, Ackermann L, Hoppe T. Ring of Change: CDC48/p97 Drives Protein Dynamics at Chromatin. Front Genet 2016; 7:73. [PMID: 27200082 PMCID: PMC4853748 DOI: 10.3389/fgene.2016.00073] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/16/2016] [Indexed: 12/31/2022] Open
Abstract
The dynamic composition of proteins associated with nuclear DNA is a fundamental property of chromosome biology. In the chromatin compartment dedicated protein complexes govern the accurate synthesis and repair of the genomic information and define the state of DNA compaction in vital cellular processes such as chromosome segregation or transcription. Unscheduled or faulty association of protein complexes with DNA has detrimental consequences on genome integrity. Consequently, the association of protein complexes with DNA is remarkably dynamic and can respond rapidly to cellular signaling events, which requires tight spatiotemporal control. In this context, the ring-like AAA+ ATPase CDC48/p97 emerges as a key regulator of protein complexes that are marked with ubiquitin or SUMO. Mechanistically, CDC48/p97 functions as a segregase facilitating the extraction of substrate proteins from the chromatin. As such, CDC48/p97 drives molecular reactions either by directed disassembly or rearrangement of chromatin-bound protein complexes. The importance of this mechanism is reflected by human pathologies linked to p97 mutations, including neurodegenerative disorders, oncogenesis, and premature aging. This review focuses on the recent insights into molecular mechanisms that determine CDC48/p97 function in the chromatin environment, which is particularly relevant for cancer and aging research.
Collapse
Affiliation(s)
- André Franz
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Institute for Genetics, University of Cologne Cologne, Germany
| | - Leena Ackermann
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Institute for Genetics, University of Cologne Cologne, Germany
| | - Thorsten Hoppe
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Institute for Genetics, University of Cologne Cologne, Germany
| |
Collapse
|
39
|
Masoumi KC, Marfany G, Wu Y, Massoumi R. Putative role of SUMOylation in controlling the activity of deubiquitinating enzymes in cancer. Future Oncol 2016; 12:565-74. [PMID: 26777062 DOI: 10.2217/fon.15.320] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Deubiquitinating enzymes (DUBs) are specialized proteins that can recognize ubiquitinated proteins, and after direct interaction, deconjugate monomeric or polymeric ubiquitin chains, thus changing the fate of the substrates. This process is instrumental in mediating or changing downstream signaling pathways. Beside mutations and alterations in their expression levels, the activity and stability of deubiquitinating enzymes is vital for their function. SUMOylations consist of the conjugation of the small peptide SUMO to protein substrates which is very similar to ubiquitination in the mechanistic and machinery required. In this review, we will focus on how SUMOylation can regulate DUB enzymatic activity, stability or DUB interaction with partners and substrates, in cancer. Furthermore, we will discuss the impact of these recent findings in the identification of new potential tools for efficient anticancer treatment strategies.
Collapse
Affiliation(s)
- Katarzyna C Masoumi
- Department of Laboratory Medicine, Medicon Village, Lund University, 22381 Lund, Sweden
| | - Gemma Marfany
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain.,Institut de Biomedicina (IBUB), Universitat de Barcelona, 08007 Barcelona, Spain.,CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Yingli Wu
- Department of Pathophysiology, Chemical Biology Division of Shanghai Universities E-Institutes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ramin Massoumi
- Department of Laboratory Medicine, Medicon Village, Lund University, 22381 Lund, Sweden
| |
Collapse
|
40
|
Anderson DJ, Le Moigne R, Djakovic S, Kumar B, Rice J, Wong S, Wang J, Yao B, Valle E, Kiss von Soly S, Madriaga A, Soriano F, Menon MK, Wu ZY, Kampmann M, Chen Y, Weissman JS, Aftab BT, Yakes FM, Shawver L, Zhou HJ, Wustrow D, Rolfe M. Targeting the AAA ATPase p97 as an Approach to Treat Cancer through Disruption of Protein Homeostasis. Cancer Cell 2015; 28:653-665. [PMID: 26555175 PMCID: PMC4941640 DOI: 10.1016/j.ccell.2015.10.002] [Citation(s) in RCA: 291] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 08/04/2015] [Accepted: 10/06/2015] [Indexed: 02/06/2023]
Abstract
p97 is a AAA-ATPase with multiple cellular functions, one of which is critical regulation of protein homeostasis pathways. We describe the characterization of CB-5083, a potent, selective, and orally bioavailable inhibitor of p97. Treatment of tumor cells with CB-5083 leads to accumulation of poly-ubiquitinated proteins, retention of endoplasmic reticulum-associated degradation (ERAD) substrates, and generation of irresolvable proteotoxic stress, leading to activation of the apoptotic arm of the unfolded protein response. In xenograft models, CB-5083 causes modulation of key p97-related pathways, induces apoptosis, and has antitumor activity in a broad range of both hematological and solid tumor models. Molecular determinants of CB-5083 activity include expression of genes in the ERAD pathway, providing a potential strategy for patient selection.
Collapse
Affiliation(s)
| | | | | | | | - Julie Rice
- Cleave Biosciences, Inc., Burlingame, CA 94010, USA
| | - Steve Wong
- Cleave Biosciences, Inc., Burlingame, CA 94010, USA
| | - Jinhai Wang
- Cleave Biosciences, Inc., Burlingame, CA 94010, USA
| | - Bing Yao
- Cleave Biosciences, Inc., Burlingame, CA 94010, USA
| | | | | | | | | | | | - Zhi Yong Wu
- Cleave Biosciences, Inc., Burlingame, CA 94010, USA
| | - Martin Kampmann
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuwen Chen
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jonathan S Weissman
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Blake T Aftab
- Division of Hematology & Oncology, Department of Medicine, Helen Diller Family Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | | | - Han-Jie Zhou
- Cleave Biosciences, Inc., Burlingame, CA 94010, USA
| | | | - Mark Rolfe
- Cleave Biosciences, Inc., Burlingame, CA 94010, USA
| |
Collapse
|
41
|
Schweitzer K, Pralow A, Naumann M. p97/VCP promotes Cullin-RING-ubiquitin-ligase/proteasome-dependent degradation of IκBα and the preceding liberation of RelA from ubiquitinated IκBα. J Cell Mol Med 2015; 20:58-70. [PMID: 26463447 PMCID: PMC4717852 DOI: 10.1111/jcmm.12702] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 08/25/2015] [Indexed: 12/21/2022] Open
Abstract
Cullin‐RING‐ubiquitin‐ligase (CRL)‐dependent ubiquitination of the nuclear factor kappa B (NF‐κB) inhibitor IκBα and its subsequent degradation by the proteasome usually precede NF‐κB/RelA nuclear activity. Through removal of the CRL‐activating modification of their cullin subunit with the ubiquitin (Ub)‐like modifier NEDD8, the COP9 signalosome (CSN) opposes CRL Ub‐ligase activity. While RelA phosphorylation was observed to mediate NF‐κB activation independent of Ub‐proteasome‐pathway (UPP)‐dependent turnover of IκBα in some studies, a strict requirement of the p97/VCP ATPase for both, IκBα degradation and NF‐κB activation, was reported in others. In this study, we thus aimed to reconcile the mechanism for tumour necrosis factor (TNF)‐induced NF‐κB activation. We found that inducible phosphorylation of RelA is accomplished in an IKK‐complex‐dependent manner within the NF‐κB/RelA‐IκBα‐complex contemporaneous with the phosphorylation of IκBα, and that RelA phosphorylation is not sufficient to dissociate NF‐κB/RelA from IκBα. Subsequent to CRL‐dependent IκBα ubiquitination functional p97/VCP is essentially required for efficient liberation of (phosphorylated) RelA from IκBα, preceding p97/VCP‐promoted timely and efficient degradation of IκBα as well as simultaneous NF‐κB/RelA nuclear translocation. Collectively, our data add new facets to the knowledge about maintenance of IκBα and RelA expression, likely depending on p97/VCP‐supported scheduled basal NF‐κB activity, and the mechanism of TNF‐induced NF‐κB activation.
Collapse
Affiliation(s)
- Katrin Schweitzer
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - Alexander Pralow
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| |
Collapse
|
42
|
Tao S, Tillotson J, Wijeratne EMK, Xu YM, Kang M, Wu T, Lau EC, Mesa C, Mason DJ, Brown RV, Clair JJL, Gunatilaka AAL, Zhang DD, Chapman E. Withaferin A Analogs That Target the AAA+ Chaperone p97. ACS Chem Biol 2015; 10:1916-1924. [PMID: 26006219 PMCID: PMC4593394 DOI: 10.1021/acschembio.5b00367] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Understanding the mode of action (MOA) of many natural products can be puzzling with mechanistic clues that seem to lack a common thread. One such puzzle lies in the evaluation of the antitumor properties of the natural product withaferin A (WFA). A variety of seemingly unrelated pathways have been identified to explain its activity, suggesting a lack of selectivity. We now show that WFA acts as an inhibitor of the chaperone, p97, both in vitro and in cell models in addition to inhibiting the proteasome in vitro. Through medicinal chemistry, we have refined the activity of WFA toward p97 and away from the proteasome. Subsequent studies indicated that these WFA analogs retained p97 activity and cytostatic activity in cell models, suggesting that the modes of action reported for WFA could be connected by proteostasis modulation. Through this endeavor, we highlight how the parallel integration of medicinal chemistry with chemical biology offers a potent solution to one of natures' intriguing molecular puzzles.
Collapse
Affiliation(s)
- Shasha Tao
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 1703 East Mabel Street, P.O. Box 210207, Tucson, Arizona 85721, United States
| | - Joseph Tillotson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 1703 East Mabel Street, P.O. Box 210207, Tucson, Arizona 85721, United States
| | - E. M. Kithsiri Wijeratne
- Southwest Center for Natural Products Research and Commercialization, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, University of Arizona, 250 E. Valencia Road, Tucson, Arizona 85706, United States
| | - Ya-ming Xu
- Southwest Center for Natural Products Research and Commercialization, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, University of Arizona, 250 E. Valencia Road, Tucson, Arizona 85706, United States
| | - MinJin Kang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 1703 East Mabel Street, P.O. Box 210207, Tucson, Arizona 85721, United States
| | - Tongde Wu
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 1703 East Mabel Street, P.O. Box 210207, Tucson, Arizona 85721, United States
| | - Eric C. Lau
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 1703 East Mabel Street, P.O. Box 210207, Tucson, Arizona 85721, United States
| | - Celestina Mesa
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 1703 East Mabel Street, P.O. Box 210207, Tucson, Arizona 85721, United States
| | - Damian J. Mason
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 1703 East Mabel Street, P.O. Box 210207, Tucson, Arizona 85721, United States
| | - Robert V. Brown
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 1703 East Mabel Street, P.O. Box 210207, Tucson, Arizona 85721, United States
| | - James J. La Clair
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 1703 East Mabel Street, P.O. Box 210207, Tucson, Arizona 85721, United States
| | - A. A. Leslie Gunatilaka
- Southwest Center for Natural Products Research and Commercialization, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, University of Arizona, 250 E. Valencia Road, Tucson, Arizona 85706, United States
| | - Donna D. Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 1703 East Mabel Street, P.O. Box 210207, Tucson, Arizona 85721, United States,Corresponding Authors.
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 1703 East Mabel Street, P.O. Box 210207, Tucson, Arizona 85721, United States,Corresponding Authors.
| |
Collapse
|
43
|
Cui Y, Niu M, Zhang X, Zhong Z, Wang J, Pang D. High expression of valosin-containing protein predicts poor prognosis in patients with breast carcinoma. Tumour Biol 2015; 36:9919-27. [PMID: 26168958 DOI: 10.1007/s13277-015-3748-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/02/2015] [Indexed: 02/03/2023] Open
Abstract
Valosin-containing protein (VCP) is one of the AAA-ATPase superfamily members. The correlation between elevated expression of VCP and progression, prognosis, and the metastatic potential has been identified in various tumor types. However, the clinical impact of VCP in breast carcinoma has not been investigated. In the current study, the expression of VCP in 421 breast tumors and adjacent normal breast tissues was examined to investigate the correlation between VCP expression and clinicopathological features in patients with breast carcinoma. We found that the expression of VCP correlated with the TNM stage, Ki67 labeling, and lymph node metastasis (LNM). The expression of VCP was increased significantly in the cytoplasm of cancer cells compared to normal mammary epithelial cells, which was associated with decreased overall survival rates of patients with breast carcinoma (P < 0.001). In conclusion, this study demonstrates significant correlation between the cytoplasmic expression of VCP and adverse prognosis in breast carcinoma, suggesting that VCP may serve as a prognostic biomarker in breast carcinoma.
Collapse
Affiliation(s)
- Yan Cui
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Haping Road No.150, Nangang District, Harbin, 150081, Heilongjiang Province, China
| | - Ming Niu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Haping Road No.150, Nangang District, Harbin, 150081, Heilongjiang Province, China
| | - Xianyu Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Haping Road No.150, Nangang District, Harbin, 150081, Heilongjiang Province, China
| | - Zhenbin Zhong
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Haping Road No.150, Nangang District, Harbin, 150081, Heilongjiang Province, China
| | - Ji Wang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Haping Road No.150, Nangang District, Harbin, 150081, Heilongjiang Province, China
| | - Da Pang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Haping Road No.150, Nangang District, Harbin, 150081, Heilongjiang Province, China.
| |
Collapse
|
44
|
Nadeau MÈ, Rico C, Tsoi M, Vivancos M, Filimon S, Paquet M, Boerboom D. Pharmacological targeting of valosin containing protein (VCP) induces DNA damage and selectively kills canine lymphoma cells. BMC Cancer 2015; 15:479. [PMID: 26104798 PMCID: PMC4479320 DOI: 10.1186/s12885-015-1489-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 06/14/2015] [Indexed: 12/20/2022] Open
Abstract
Background Valosin containing protein (VCP) is a critical mediator of protein homeostasis and may represent a valuable therapeutic target for several forms of cancer. Overexpression of VCP occurs in many cancers, and often in a manner correlating with malignancy and poor outcome. Here, we analyzed VCP expression in canine lymphoma and assessed its potential as a therapeutic target for this disease. Methods VCP expression in canine lymphomas was evaluated by immunoblotting and immunohistochemistry. The canine lymphoma cell lines CLBL-1, 17–71 and CL-1 were treated with the VCP inhibitor Eeyarestatin 1 (EER-1) at varying concentrations and times and were assessed for viability by trypan blue exclusion, apoptosis by TUNEL and caspase activity assays, and proliferation by propidium iodide incorporation and FACS. The mechanism of EER-1 action was determined by immunoblotting and immunofluorescence analyses of Lys48 ubiquitin and markers of ER stress (DDIT3), autophagy (SQSTM1, MAP1LC3A) and DNA damage (γH2AFX). TRP53/ATM-dependent signaling pathway activity was assessed by immunoblotting for TRP53 and phospho-TRP53 and real-time RT-PCR measurement of Cdkn1a mRNA. Results VCP expression levels in canine B cell lymphomas were found to increase with grade. EER-1 treatment killed canine lymphoma cells preferentially over control peripheral blood mononuclear cells. EER-1 treatment of CLBL-1 cells was found to both induce apoptosis and cell cycle arrest in G1. Unexpectedly, EER-1 did not appear to act either by inducing ER stress or inhibiting the aggresome-autophagy pathway. Rather, a rapid and dramatic increase in γH2AFX expression was noted, indicating that EER-1 may act by promoting DNA damage accumulation. Increased TRP53 phosphorylation and Cdkn1a mRNA levels indicated an activation of the TRP53/ATM DNA damage response pathway in response to EER-1, likely contributing to the induction of apoptosis and cell cycle arrest. Conclusions These results correlate VCP expression with malignancy in canine B cell lymphoma. The selective activity of EER-1 against lymphoma cells suggests that VCP will represent a clinically useful therapeutic target for the treatment of lymphoma. We further suggest a mechanism of EER-1 action centered on the DNA repair response that may be of central importance for the design and characterization of VCP inhibitory compounds for therapeutic use. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1489-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marie-Ève Nadeau
- Département des Sciences Cliniques, Université de Montréal, Saint-Hyacinthe, QC, J2S7C6, Canada.
| | - Charlène Rico
- Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, J2S7C6, Canada.
| | - Mayra Tsoi
- Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, J2S7C6, Canada.
| | - Mélanie Vivancos
- Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, J2S7C6, Canada.
| | - Sabin Filimon
- Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, J2S7C6, Canada.
| | - Marilène Paquet
- Département de Pathologie et de Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, J2S7C6, Canada.
| | - Derek Boerboom
- Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, J2S7C6, Canada.
| |
Collapse
|
45
|
Chapman E, Maksim N, de la Cruz F, La Clair JJ. Inhibitors of the AAA+ chaperone p97. Molecules 2015; 20:3027-49. [PMID: 25685910 PMCID: PMC4576884 DOI: 10.3390/molecules20023027] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/03/2015] [Indexed: 12/22/2022] Open
Abstract
It is remarkable that a pathway as ubiquitous as protein quality control can be targeted to treat cancer. Bortezomib, an inhibitor of the proteasome, was first approved by the US Food and Drug Administration (FDA) more than 10 years ago to treat refractory myeloma and later extended to lymphoma. Its use has increased the survival rate of myeloma patients by as much as three years. This success was followed with the recent accelerated approval of the natural product derived proteasome inhibitor carfilzomib (Kyprolis®), which is used to treat patients with bortezomib-resistant multiple myeloma. The success of these two drugs has validated protein quality control as a viable target to fight select cancers, but begs the question why are proteasome inhibitors limited to lymphoma and myeloma? More recently, these limitations have encouraged the search for additional targets within the protein quality control system that might offer heightened cancer cell specificity, enhanced clinical utility, a lower rate of resistance, reduced toxicity, and mitigated side effects. One promising target is p97, an ATPase associated with various cellular activities (AAA+) chaperone. p97 figures prominently in protein quality control as well as serving a variety of other cellular functions associated with cancer. More than a decade ago, it was determined that up-regulation of p97 in many forms of cancer correlates with a poor clinical outcome. Since these initial discoveries, a mechanistic explanation for this observation has been partially illuminated, but details are lacking. Understandably, given this clinical correlation, myriad roles within the cell, and its importance in protein quality control, p97 has emerged as a potential therapeutic target. This review provides an overview of efforts towards the discovery of small molecule inhibitors of p97, offering a synopsis of efforts that parallel the excellent reviews that currently exist on p97 structure, function, and physiology.
Collapse
Affiliation(s)
- Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721-0207, USA.
| | - Nick Maksim
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721-0207, USA.
| | - Fabian de la Cruz
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721-0207, USA.
| | - James J La Clair
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721-0207, USA.
| |
Collapse
|
46
|
Marza E, Taouji S, Barroso K, Raymond AA, Guignard L, Bonneu M, Pallares-Lupon N, Dupuy JW, Fernandez-Zapico ME, Rosenbaum J, Palladino F, Dupuy D, Chevet E. Genome-wide screen identifies a novel p97/CDC-48-dependent pathway regulating ER-stress-induced gene transcription. EMBO Rep 2015; 16:332-40. [PMID: 25652260 DOI: 10.15252/embr.201439123] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The accumulation of misfolded proteins in the endoplasmic reticulum (ER) activates the Unfolded Protein Response (UPR(ER)) to restore ER homeostasis. The AAA(+) ATPase p97/CDC-48 plays key roles in ER stress by promoting both ER protein degradation and transcription of UPR(ER) genes. Although the mechanisms associated with protein degradation are now well established, the molecular events involved in the regulation of gene transcription by p97/CDC-48 remain unclear. Using a reporter-based genome-wide RNAi screen in combination with quantitative proteomic analysis in Caenorhabditis elegans, we have identified RUVB-2, a AAA(+) ATPase, as a novel repressor of a subset of UPR(ER) genes. We show that degradation of RUVB-2 by CDC-48 enhances expression of ER stress response genes through an XBP1-dependent mechanism. The functional interplay between CDC-48 and RUVB-2 in controlling transcription of select UPR(ER) genes appears conserved in human cells. Together, these results describe a novel role for p97/CDC-48, whereby its role in protein degradation is integrated with its role in regulating expression of ER stress response genes.
Collapse
Affiliation(s)
- Esther Marza
- Team "Endoplasmic Reticulum stress and cancer", INSERM, UMR1053, Bordeaux, France University of Bordeaux, Bordeaux, France ARNA laboratory, INSERM U869, Bordeaux, France
| | - Saïd Taouji
- Team "Endoplasmic Reticulum stress and cancer", INSERM, UMR1053, Bordeaux, France University of Bordeaux, Bordeaux, France
| | - Kim Barroso
- Team "Endoplasmic Reticulum stress and cancer", INSERM, UMR1053, Bordeaux, France University of Bordeaux, Bordeaux, France
| | - Anne-Aurélie Raymond
- University of Bordeaux, Bordeaux, France "REPTeam", INSERM, UMR1053, Bordeaux, France
| | - Léo Guignard
- University of Bordeaux, Bordeaux, France ARNA laboratory, INSERM U869, Bordeaux, France
| | - Marc Bonneu
- University of Bordeaux, Bordeaux, France Plateforme Proteome, Bordeaux, France
| | - Néstor Pallares-Lupon
- Team "Endoplasmic Reticulum stress and cancer", INSERM, UMR1053, Bordeaux, France University of Bordeaux, Bordeaux, France
| | - Jean-William Dupuy
- University of Bordeaux, Bordeaux, France Plateforme Proteome, Bordeaux, France
| | - Martin E Fernandez-Zapico
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, MN, USA
| | - Jean Rosenbaum
- University of Bordeaux, Bordeaux, France "REPTeam", INSERM, UMR1053, Bordeaux, France
| | - Francesca Palladino
- Laboratory of Molecular and Cellular Biology, Ecole Normale Supérieure, CNRS UMR5239 Université de Lyon, Lyon Cedex 07, France
| | - Denis Dupuy
- University of Bordeaux, Bordeaux, France ARNA laboratory, INSERM U869, Bordeaux, France
| | - Eric Chevet
- Team "Endoplasmic Reticulum stress and cancer", INSERM, UMR1053, Bordeaux, France University of Bordeaux, Bordeaux, France Centre Régional de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| |
Collapse
|
47
|
Abstract
The clinical successes of proteasome inhibitors for the treatment of cancer have highlighted the therapeutic potential of targeting this protein degradation system. However, proteasome inhibitors prevent the degradation of numerous proteins, which may cause adverse effects. Increased specificity could be achieved by inhibiting the components of the ubiquitin-proteasome system that target specific subsets of proteins for degradation. F-box proteins are the substrate-targeting subunits of SKP1-CUL1-F-box protein (SCF) ubiquitin ligase complexes. Through the degradation of a plethora of diverse substrates, SCF ubiquitin ligases control a multitude of processes at the cellular and organismal levels, and their dysregulation is implicated in many pathologies. SCF ubiquitin ligases are characterized by their high specificity for substrates, and these ligases therefore represent promising drug targets. However, the potential for therapeutic manipulation of SCF complexes remains an underdeveloped area. This Review explores and discusses potential strategies to target SCF-mediated biological processes to treat human diseases.
Collapse
Affiliation(s)
- Jeffrey R Skaar
- 1] Department of Pathology, Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, 522 First Avenue, SRB 1107, New York, New York 10016, USA. [2]
| | - Julia K Pagan
- 1] Department of Pathology, Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, 522 First Avenue, SRB 1107, New York, New York 10016, USA. [2]
| | - Michele Pagano
- 1] Department of Pathology, Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, 522 First Avenue, SRB 1107, New York, New York 10016, USA. [2] Howard Hughes Medical Institute
| |
Collapse
|
48
|
Abstract
Genomic alterations may make cancer cells more dependent than normal cells on mechanisms of proteostasis, including protein folding and degradation. This proposition is the basis for the clinical use of proteasome inhibitors to treat multiple myeloma and mantle cell lymphoma. However, proteasome inhibitors have not proved effective in treating other cancers, and this has called into question the general applicability of this approach. Here, I consider possible explanations for this apparently limited applicability, and discuss whether inhibiting other broadly acting components of the ubiquitin-proteasome system - including ubiquitin-activating enzyme and the AAA-ATPase p97/VCP - might be more generally effective in cancer therapy.
Collapse
Affiliation(s)
- Raymond J Deshaies
- Division of Biology and Biological Engineering and Howard Hughes Medical Institute, California Institute of Technology, Pasadena 91107, CA, USA.
| |
Collapse
|
49
|
Zhao X, Wu Y, Duan J, Ma Y, Shen Z, Wei L, Cui X, Zhang J, Xie Y, Liu J. Quantitative Proteomic Analysis of Exosome Protein Content Changes Induced by Hepatitis B Virus in Huh-7 Cells Using SILAC Labeling and LC–MS/MS. J Proteome Res 2014; 13:5391-402. [PMID: 25265333 DOI: 10.1021/pr5008703] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Xue Zhao
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
- Microbiology
Laboratory, Shanghai Municipal Center for Disease Control and Prevention, No. 1380 West Zhongshan Road, Shanghai 200336, People’s Republic of China
| | - Yanxin Wu
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
| | - Jinlin Duan
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
- Department
of Pathology, Tongren Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 786 YuYuan Road, Shanghai 200336, People’s Republic of China
| | - Yanchun Ma
- Lab
Center, Putuo District Center Hospital, Shanghai University of Traditional Chinese Medicine, 164 Lanxi Road, Shanghai 200062, People’s Republic of China
| | - Zhongliang Shen
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
| | - Lili Wei
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
| | - Xiaoxian Cui
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
| | - Junqi Zhang
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
| | - Youhua Xie
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
| | - Jing Liu
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
| |
Collapse
|
50
|
Kang M, Wu T, Wijeratne EMK, Lau EC, Mason DJ, Mesa C, Tillotson J, Zhang DD, Gunatilaka AAL, La Clair JJ, Chapman E. Functional chromatography reveals three natural products that target the same protein with distinct mechanisms of action. Chembiochem 2014; 15:2125-31. [PMID: 25125376 PMCID: PMC4187115 DOI: 10.1002/cbic.201402258] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Indexed: 01/12/2023]
Abstract
Access to lead compounds with defined molecular targets continues to be a barrier to the translation of natural product resources. As a solution, we developed a system that uses discrete, recombinant proteins as the vehicles for natural product isolation. Here, we describe the use of this functional chromatographic method to identify natural products that bind to the AAA+ chaperone, p97, a promising cancer target. Application of this method to a panel of fungal and plant extracts identified rheoemodin, 1-hydroxydehydroherbarin, and phomapyrrolidone A as distinct p97 modulators. Excitingly, each of these molecules displayed a unique mechanism of p97 modulation. This discovery provides strong support for the application of functional chromatography to the discovery of protein modulators that would likely escape traditional high-throughput or phenotypic screening platforms.
Collapse
Affiliation(s)
- MinJin Kang
- College of Pharmacy, Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721-0207, United States
| | - Tongde Wu
- College of Pharmacy, Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721-0207, United States
| | - E. M. Kithsiri Wijeratne
- Southwest Center for Natural Products Research and Commercialization, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ 85706-6800, United States
| | - Eric C. Lau
- College of Pharmacy, Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721-0207, United States
| | - Damian J. Mason
- College of Pharmacy, Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721-0207, United States
| | - Celestina Mesa
- College of Pharmacy, Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721-0207, United States
| | - Joseph Tillotson
- College of Pharmacy, Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721-0207, United States
| | - Donna D. Zhang
- College of Pharmacy, Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721-0207, United States
| | - A. A. Leslie Gunatilaka
- Southwest Center for Natural Products Research and Commercialization, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ 85706-6800, United States
| | - James J. La Clair
- Xenobe Research Institute, P. O. Box 3052, San Diego, CA 92163-1052, United States
| | - Eli Chapman
- College of Pharmacy, Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721-0207, United States
| |
Collapse
|