1
|
Guo Q, Qin H, Chen Z, Zhang W, Zheng L, Qin T. Key roles of ubiquitination in regulating critical regulators of cancer stem cell functionality. Genes Dis 2025; 12:101311. [PMID: 40034124 PMCID: PMC11875185 DOI: 10.1016/j.gendis.2024.101311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/23/2024] [Accepted: 03/07/2024] [Indexed: 03/05/2025] Open
Abstract
The ubiquitin (Ub) system, a ubiquitous presence across eukaryotes, plays a crucial role in the precise orchestration of diverse cellular protein processes. From steering cellular signaling pathways and orchestrating cell cycle progression to guiding receptor trafficking and modulating immune responses, this process plays a crucial role in regulating various biological functions. The dysregulation of Ub-mediated signaling pathways in prevalent cancers ushers in a spectrum of clinical outcomes ranging from tumorigenesis and metastasis to recurrence and drug resistance. Ubiquitination, a linchpin process mediated by Ub, assumes a central mantle in molding cellular signaling dynamics. It navigates transitions in biological cues and ultimately shapes the destiny of proteins. Recent years have witnessed an upsurge in the momentum surrounding the development of protein-based therapeutics aimed at targeting the Ub system under the sway of cancer stem cells. The article provides a comprehensive overview of the ongoing in-depth discussions regarding the regulation of the Ub system and its impact on the development of cancer stem cells. Amidst the tapestry of insights, the article delves into the expansive roles of E3 Ub ligases, deubiquitinases, and transcription factors entwined with cancer stem cells. Furthermore, the spotlight turns to the interplay with pivotal signaling pathways the Notch, Hedgehog, Wnt/β-catenin, and Hippo-YAP signaling pathways all play crucial roles in the regulation of cancer stem cells followed by the specific modulation of Ub-proteasome.
Collapse
Affiliation(s)
- Qianqian Guo
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Hai Qin
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, Guizhou 550014, China
| | - Zelong Chen
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Artificial Intelligence and IoT Smart Medical Engineering Research Center of Henan Province, Zhengzhou, Henan 450008, China
| | - Wenzhou Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Lufeng Zheng
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Tingting Qin
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| |
Collapse
|
2
|
Roy A, Sharma S, Paul I, Ray S. Molecular hybridization assisted multi-technique approach for designing USP21 inhibitors to halt catalytic triad-mediated nucleophilic attack and suppress pancreatic ductal adenocarcinoma progression: A molecular dynamics study. Comput Biol Med 2024; 182:109096. [PMID: 39270458 DOI: 10.1016/j.compbiomed.2024.109096] [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: 12/17/2023] [Revised: 07/20/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024]
Abstract
AIMS Pancreatic cancer, the 12th-most common cancer, globally, is highly challenging to treat due to its complex epigenetic, metabolic, and genomic characteristics. In pancreatic ductal adenocarcinoma, USP21 acts as an oncogene by stabilizing the long isoform of Transcription Factor 7, thereby activating the Wnt signaling pathway. This study aims to inhibit activation of this pathway through computer-aided drug discovery. Accordingly, four libraries of compounds were designed to target the USP21's catalytic domain (Cys221, His518, Asp534), responsible for its deubiquitinating activity. MAIN METHODS Utilizing an array of computer-aided drug design methodologies, such as molecular docking, virtual screening, principal component analysis, molecular dynamics simulation, and dynamic cross-correlation matrix, the structural and functional characteristics of the USP21-inhibitor complex were examined. Following the evaluation of the binding affinities, 20 potential ligands were selected, and the best ligand was subjected to additional molecular dynamics simulation study. KEY FINDINGS The results indicated that the ligand-bound USP21 exhibited reduced structural fluctuations compared to the unbound form, as evident from RMSD, RMSF, Rg, and SASA graphs. ADMET analysis of the top ligand showed promising pharmacokinetic and pharmacodynamic profiles, good bioavailability, and low toxicity. The stable conformations of the proposed drug when bound to their target cavities indicate a robust binding affinity of -9.3 kcal/mol. The drug exhibits an elevated pKi value of 6.82, a noteworthy pIC50 value of 5.972, and a pKd value of 6.023 proving its high affinity and inhibitory potential towards the target. SIGNIFICANCE In-vitro testing of the top compound (MOLHYB-0436) could lead to its use as a potential treatment for pancreatic cancer.
Collapse
Affiliation(s)
- Alankar Roy
- Amity Institute of Biotechnology, Amity University, Kolkata, India
| | - Sayan Sharma
- Amity Institute of Biotechnology, Amity University, Kolkata, India
| | - Ishani Paul
- Amity Institute of Biotechnology, Amity University, Kolkata, India
| | - Sujay Ray
- Amity Institute of Biotechnology, Amity University, Kolkata, India.
| |
Collapse
|
3
|
Yang D, Liu X, Yang Y, Long Y, Nan D, Shi B, Wang J, Yang M, Cong H, Xing L, Zhou F, Yuan Q, Ta N, Zhang Y, Ma R, Liu F, Liu S. Pharmacological USP2 targeting suppresses ovarian cancer growth by potentiating apoptosis and ferroptosis. Arch Biochem Biophys 2024; 762:110193. [PMID: 39486565 DOI: 10.1016/j.abb.2024.110193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/27/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Ovarian cancer is a frequently observed type of gynaecologic malignancy generally associated with poor prognosis around the world. Ubiquitin-specific proteases (USPs) form the largest subfamily of deubiquitylating enzymes and have emerged as potential therapeutic targets against human cancers. Through a systematic analysis of the prognostic significance of USP expression, USP2 was found to be inversely correlated with patient survival in ovarian cancer. Accordingly, we investigated the effects of pharmacological inhibition of USP2 on ovarian cancer by exploiting its small molecule inhibitor ML364. Our findings show that ML364 effectively hindered ovarian cancer growth and migration using a series of in vitro assays. In addition to apoptosis induction, ML364 also sensitized ovarian cancer cells to ferroptosis. Mechanistically, ML364 treatment resulted in cyclin D1 downregulation, increased poly (ADP-ribose) polymerase (PARP) cleavage, and elevated ROS levels in ovarian cancer cells. Collectively, our findings suggest USP2 as a potential therapeutic target in ovarian cancer, and hence, its pharmacological inhibition warrants further investigation.
Collapse
Affiliation(s)
- Dian Yang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xiuxiu Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China; Department of Gynecology, Zhongshan Hospital of Dalian University, Dalian, China
| | - Yinghui Yang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yu Long
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Ding Nan
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Bo Shi
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Jinhao Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Mei Yang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Haotian Cong
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Lin Xing
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Feixue Zhou
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Qianhui Yuan
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Na Ta
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yingqiu Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Ruilan Ma
- Department of Radiation Oncology, The Second Affiliated Hospital, Dalian Medical University, China.
| | - Fang Liu
- Department of Oncology, The Second Affiliated Hospital, Dalian Medical University, China.
| | - Shuyan Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
| |
Collapse
|
4
|
Jamal A. E3 Ubiquitin Ligases and Their Therapeutic Applications in Cancers: Narrative Review. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2024; 16:S1984-S1986. [PMID: 39346323 PMCID: PMC11426868 DOI: 10.4103/jpbs.jpbs_134_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 10/01/2024] Open
Abstract
E3 ubiquitin ligases are a class of enzymes, essential for maintaining the equilibrium of cells by binding ubiquitin molecules to substrates to mark them for destruction. Since many cancer-related proteins, including both oncogenic and tumor-suppressive ones, are controlled by the ubiquitin-proteasome system, E3 ligases have drawn a great deal of interest as potential targets for the creation of anti-cancer drugs. This is because E3 ligases function as modules that select the substrates that are intended for degradation, giving them the ability to particularly affect proteins that are connected to cancer. Their molecular properties and the ways in which they work serve as the basis for these distinctions. Investment in the creation of bioactive substances that can target E3 ligases is essential given the crucial roles they play in cancer. These substances have the potential to be powerful cancer-fighting tools. In this review, we explore the crucial roles that E3 ligases play in the biology of cancer. We also examine the current bioactive substances that have been created to target different E3 ligases, emphasizing their potential as candidates for treating the cancers.
Collapse
Affiliation(s)
- Azfar Jamal
- Department of Biology, College of Science, Al-Zulfi-, Majmaah University, Majmaah, Riyadh, Saudi Arabia
- Health and Basic Science Research Centre, Majmaah University, Majmaah, Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Lospinoso Severini L, Loricchio E, Navacci S, Basili I, Alfonsi R, Bernardi F, Moretti M, Conenna M, Cucinotta A, Coni S, Petroni M, De Smaele E, Giannini G, Maroder M, Canettieri G, Mastronuzzi A, Guardavaccaro D, Ayrault O, Infante P, Bufalieri F, Di Marcotullio L. SALL4 is a CRL3 REN/KCTD11 substrate that drives Sonic Hedgehog-dependent medulloblastoma. Cell Death Differ 2024; 31:170-187. [PMID: 38062245 PMCID: PMC10850099 DOI: 10.1038/s41418-023-01246-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 02/09/2024] Open
Abstract
The Sonic Hedgehog (SHH) pathway is crucial regulator of embryonic development and stemness. Its alteration leads to medulloblastoma (MB), the most common malignant pediatric brain tumor. The SHH-MB subgroup is the best genetically characterized, however the molecular mechanisms responsible for its pathogenesis are not fully understood and therapeutic benefits are still limited. Here, we show that the pro-oncogenic stemness regulator Spalt-like transcriptional factor 4 (SALL4) is re-expressed in mouse SHH-MB models, and its high levels correlate with worse overall survival in SHH-MB patients. Proteomic analysis revealed that SALL4 interacts with REN/KCTD11 (here REN), a substrate receptor subunit of the Cullin3-RING ubiquitin ligase complex (CRL3REN) and a tumor suppressor lost in ~30% of human SHH-MBs. We demonstrate that CRL3REN induces polyubiquitylation and degradation of wild type SALL4, but not of a SALL4 mutant lacking zinc finger cluster 1 domain (ΔZFC1). Interestingly, SALL4 binds GLI1 and cooperates with HDAC1 to potentiate GLI1 deacetylation and transcriptional activity. Notably, inhibition of SALL4 suppresses SHH-MB growth both in murine and patient-derived xenograft models. Our findings identify SALL4 as a CRL3REN substrate and a promising therapeutic target in SHH-dependent cancers.
Collapse
Affiliation(s)
| | - Elena Loricchio
- Department of Molecular Medicine, University of Rome La Sapienza, 00161, Rome, Italy
| | - Shirin Navacci
- Department of Molecular Medicine, University of Rome La Sapienza, 00161, Rome, Italy
| | - Irene Basili
- Department of Molecular Medicine, University of Rome La Sapienza, 00161, Rome, Italy
- Institut Curie, PSL Research University, CNRS UMR, INSERM, 91401, Orsay, France
| | - Romina Alfonsi
- Centro Nazionale per il Controllo e la Valutazione dei Farmaci, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Flavia Bernardi
- Institut Curie, PSL Research University, CNRS UMR, INSERM, 91401, Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR, INSERM U, 91401, Orsay, France
| | - Marta Moretti
- Department of Experimental Medicine, University of Rome La Sapienza, 00161, Rome, Italy
| | - Marilisa Conenna
- Department of Molecular Medicine, University of Rome La Sapienza, 00161, Rome, Italy
| | - Antonino Cucinotta
- Department of Molecular Medicine, University of Rome La Sapienza, 00161, Rome, Italy
| | - Sonia Coni
- Department of Molecular Medicine, University of Rome La Sapienza, 00161, Rome, Italy
| | - Marialaura Petroni
- Department of Molecular Medicine, University of Rome La Sapienza, 00161, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome La Sapienza, 00161, Rome, Italy
| | - Enrico De Smaele
- Department of Experimental Medicine, University of Rome La Sapienza, 00161, Rome, Italy
| | - Giuseppe Giannini
- Department of Molecular Medicine, University of Rome La Sapienza, 00161, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome La Sapienza, 00161, Rome, Italy
| | - Marella Maroder
- Department of Molecular Medicine, University of Rome La Sapienza, 00161, Rome, Italy
| | - Gianluca Canettieri
- Department of Molecular Medicine, University of Rome La Sapienza, 00161, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome La Sapienza, 00161, Rome, Italy
| | - Angela Mastronuzzi
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | | | - Olivier Ayrault
- Institut Curie, PSL Research University, CNRS UMR, INSERM, 91401, Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR, INSERM U, 91401, Orsay, France
| | - Paola Infante
- Department of Molecular Medicine, University of Rome La Sapienza, 00161, Rome, Italy
| | - Francesca Bufalieri
- Department of Molecular Medicine, University of Rome La Sapienza, 00161, Rome, Italy.
| | - Lucia Di Marcotullio
- Department of Molecular Medicine, University of Rome La Sapienza, 00161, Rome, Italy.
- Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome La Sapienza, 00161, Rome, Italy.
| |
Collapse
|
6
|
Al-Balushi E, Al Marzouqi A, Tavoosi S, Baghsheikhi AH, Sadri A, Aliabadi LS, Salarabedi MM, Rahman SA, Al-Yateem N, Jarrahi AM, Halimi A, Ahmadvand M, Abdel-Rahman WM. Comprehensive analysis of the role of ubiquitin-specific peptidases in colorectal cancer: A systematic review. World J Gastrointest Oncol 2024; 16:197-213. [PMID: 38292842 PMCID: PMC10824112 DOI: 10.4251/wjgo.v16.i1.197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/05/2023] [Accepted: 12/07/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most frequent and the second most fatal cancer. The search for more effective drugs to treat this disease is ongoing. A better understanding of the mechanisms of CRC development and progression may reveal new therapeutic strategies. Ubiquitin-specific peptidases (USPs), the largest group of the deubiquitinase protein family, have long been implicated in various cancers. There have been numerous studies on the role of USPs in CRC; however, a comprehensive view of this role is lacking. AIM To provide a systematic review of the studies investigating the roles and functions of USPs in CRC. METHODS We systematically queried the MEDLINE (via PubMed), Scopus, and Web of Science databases. RESULTS Our study highlights the pivotal role of various USPs in several processes implicated in CRC: Regulation of the cell cycle, apoptosis, cancer stemness, epithelial-mesenchymal transition, metastasis, DNA repair, and drug resistance. The findings of this study suggest that USPs have great potential as drug targets and noninvasive biomarkers in CRC. The dysregulation of USPs in CRC contributes to drug resistance through multiple mechanisms. CONCLUSION Targeting specific USPs involved in drug resistance pathways could provide a novel therapeutic strategy for overcoming resistance to current treatment regimens in CRC.
Collapse
Affiliation(s)
- Eman Al-Balushi
- College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Amina Al Marzouqi
- College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Shima Tavoosi
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan 81746-73441, Iran
| | - Amir Hossein Baghsheikhi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran 11365/4435, Iran
| | - Arash Sadri
- Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Leyla Sharifi Aliabadi
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Mohammad-Mahdi Salarabedi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1983969411, Iran
| | - Syed Azizur Rahman
- College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Nabeel Al-Yateem
- Department of Nursing, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Alireza Mosavi Jarrahi
- Cancer Research Centre, Shahid Beheshti University of Medical Sciences, Tehran 1983969411, Iran
| | - Aram Halimi
- Cancer Research Centre, Shahid Beheshti University of Medical Sciences, Tehran 1983969411, Iran
| | - Mohammad Ahmadvand
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences , Tehran 1416634793, Iran
| | - Wael M Abdel-Rahman
- Department of Medical Laboratory Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
7
|
USP7 Inhibitors in Cancer Immunotherapy: Current Status and Perspective. Cancers (Basel) 2022; 14:cancers14225539. [PMID: 36428632 PMCID: PMC9688046 DOI: 10.3390/cancers14225539] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/03/2022] [Accepted: 11/06/2022] [Indexed: 11/12/2022] Open
Abstract
Ubiquitin-specific protease 7 (USP7) regulates the stability of a plethora of intracellular proteins involved in the suppression of anti-tumor immune responses and its overexpression is associated with poor survival in many cancers. USP7 impairs the balance of the p53/MDM2 axis resulting in the proteasomal degradation of the p53 tumor suppressor, a process that can be reversed by small-molecule inhibitors of USP7. USP7 was shown to regulate the anti-tumor immune responses in several cases. Its inhibition impedes the function of regulatory T cells, promotes polarization of tumor-associated macrophages, and reduces programmed death-ligand 1 (PD-L1) expression in tumor cells. The efficacy of small-molecule USP7 inhibitors was demonstrated in vivo. The synergistic effect of combining USP7 inhibition with cancer immunotherapy is a promising therapeutic approach, though its clinical efficacy is yet to be proven. In this review, we focus on the recent developments in understanding the intrinsic role of USP7, its interplay with other molecular pathways, and the therapeutic potential of targeting USP7 functions.
Collapse
|
8
|
Zheng W, Li S, Huang J, Dong Y, Zhang H, Zheng J. Down-Regulation of Ubiquitin-Specific Peptidase 9X Inhibited Proliferation, Migration and Invasion of Osteosarcoma <i>via</i> ERK1/2 and PI3K/Akt Signaling Pathways. Biol Pharm Bull 2022; 45:1283-1290. [DOI: 10.1248/bpb.b22-00198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Wendi Zheng
- Department of Orthopedics, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital
| | - Shuang Li
- Department of Pathology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital
| | - Jincheng Huang
- Department of Orthopedics, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital
| | - Yonghui Dong
- Department of Orthopedics, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital
| | - Hongjun Zhang
- Department of Orthopedics, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital
| | - Jia Zheng
- Department of Orthopedics, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital
| |
Collapse
|
9
|
USP7 regulates the ERK1/2 signaling pathway through deubiquitinating Raf-1 in lung adenocarcinoma. Cell Death Dis 2022; 13:698. [PMID: 35948545 PMCID: PMC9365811 DOI: 10.1038/s41419-022-05136-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 01/21/2023]
Abstract
Ubiquitin-specific protease 7 (USP7) is one of the deubiquitinating enzymes (DUBs) in the ubiquitin-specific protease (USP) family. It is a key regulator of numerous cellular functions including immune response, cell cycle, DNA damage and repair, epigenetics, and several signaling pathways. USP7 acts by removing ubiquitin from the substrate proteins. USP7 also binds to a specific binding motif of substrate proteins having the [P/A/E]-X-X-S or K-X-X-X-K protein sequences. To date, numerous substrate proteins of USP7 have been identified, but no studies have been conducted using the binding motif that USP7 binds. In the current study, we analyzed putative substrate proteins of USP7 through the [P/A/E]-X-X-S and K-X-X-X-K binding motifs using bioinformatics tools, and confirmed that Raf-1 is one of the substrates for USP7. USP7 binds to the Pro-Val-Asp-Ser (PVDS) motif of the conserved region 2 (CR2) which contains phosphorylation sites of Raf-1 and decreased M1-, K6-, K11-, K27-, K33-, and K48-linked polyubiquitination of Raf-1. We further identified that the DUB activity of USP7 decreases the threonine phosphorylation level of Raf-1 and inhibits signaling transduction through Raf activation. This regulatory mechanism inhibits the activation of the ERK1/2 signaling pathway, thereby inhibiting the G2/M transition and the cell proliferation of lung adenocarcinoma cells. In summary, our results indicate that USP7 deubiquitinates Raf-1 and is a new regulator of the ERK1/2 signaling pathway in lung adenocarcinoma.
Collapse
|
10
|
Bhattacharjee R, Ghosh S, Nath A, Basu A, Biswas O, Patil CR, Kundu CN. Theragnostic strategies harnessing the self-renewal pathways of stem-like cells in the acute myeloid leukemia. Crit Rev Oncol Hematol 2022; 177:103753. [PMID: 35803452 DOI: 10.1016/j.critrevonc.2022.103753] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 06/21/2022] [Accepted: 07/02/2022] [Indexed: 02/07/2023] Open
Abstract
Acute myelogenous leukemia (AML) is a genetically heterogeneous and aggressive cancer of the Hematopoietic Stem/progenitor cells. It is distinguished by the uncontrollable clonal growth of malignant myeloid stem cells in the bone marrow, venous blood, and other body tissues. AML is the most predominant of leukemias occurring in adults (25%) and children (15-20%). The relapse after chemotherapy is a major concern in the treatment of AML. The overall 5-year survival rate in young AML patients is about 40-45% whereas in the elderly patients it is less than 10%. Leukemia stem-like cells (LSCs) having the ability to self-renew indefinitely, repopulate and persist longer in the G0/G1 phase play a crucial role in the AML relapse and refractoriness to chemotherapy. Hence, novel treatment strategies and diagnostic biomarkers targeting LSCs are being increasingly investigated. Through this review, we have explored the signaling modulations in the LSCs as the theragnostic targets. The significance of the self-renewal pathways in overcoming the treatment challenges in AML has been highlighted.
Collapse
Affiliation(s)
- Rahul Bhattacharjee
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Sharad Ghosh
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Arijit Nath
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Asmita Basu
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Ojaswi Biswas
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Chandragauda R Patil
- Department of Pharmacology, DIPSAR, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Chanakya Nath Kundu
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India.
| |
Collapse
|
11
|
Lospinoso Severini L, Bufalieri F, Infante P, Di Marcotullio L. Proteolysis-Targeting Chimera (PROTAC): Is the Technology Looking at the Treatment of Brain Tumors? Front Cell Dev Biol 2022; 10:854352. [PMID: 35242765 PMCID: PMC8886235 DOI: 10.3389/fcell.2022.854352] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 01/31/2022] [Indexed: 12/23/2022] Open
Abstract
Post-translational modifications, such as ubiquitylation, need to be tightly controlled to guarantee the accurate localization and activity of proteins. Ubiquitylation is a dynamic process primarily responsible for proteasome-mediated degradation of substrate proteins and crucial for both normal homeostasis and disease. Alterations in ubiquitylation lead to the upregulation of oncoproteins and/or downregulation of tumor suppressors, thus concurring in tumorigenesis. PROteolysis-TArgeting Chimera (PROTAC) is an innovative strategy that takes advantage by the cell’s own Ubiquitin-Proteasome System (UPS). Each PROTAC molecule is composed by a ligand that recruits the target protein of interest (POI), a ligand specific for an E3 ubiquitin ligase enzyme, and a linker that connects these units. Upon binding to the POI, the PROTAC recruits the E3 inducing ubiquitylation-dependent proteasome degradation of the POI. To date, PROTAC technology has entered in clinical trials for several human cancers. Here, we will discuss the advantages and limitations of PROTACs development and safety considerations for their clinical application. Furthermore, we will review the potential of PROTAC strategy as therapeutic option in brain tumor, focusing on glioblastoma.
Collapse
Affiliation(s)
| | - Francesca Bufalieri
- Department of Molecular Medicine, University of Rome La Sapienza, Rome, Italy
| | - Paola Infante
- Department of Molecular Medicine, University of Rome La Sapienza, Rome, Italy
| | - Lucia Di Marcotullio
- Department of Molecular Medicine, University of Rome La Sapienza, Rome, Italy.,Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome La Sapienza, Rome, Italy
| |
Collapse
|
12
|
Huang J, Zhan Y, Jiang L, Gao Y, Zhao B, Zhang Y, Zhang W, Zheng J, Yu J. Identification of the Potential Prognosis Biomarkers in Hepatocellular Carcinoma: An Analysis Based on WGCNA and PPI. Int J Gen Med 2021; 14:9555-9565. [PMID: 34916837 PMCID: PMC8670864 DOI: 10.2147/ijgm.s338500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/25/2021] [Indexed: 12/24/2022] Open
Abstract
Aim This study was done to determine biomarkers for the prognostic prediction of hepatocellular carcinoma (HCC). Materials and Methods In the Gene Expression Omnibus, the gene expression profiles of HCC were downloaded. Biomarkers were identified by weighted gene co-expression network analysis and protein–protein interaction network analysis. Results There were 24 modules, which were characterized by the high correlation with HCC. Meanwhile, through enrichment analysis, differentially expressed genes were largely participated in the ubiquitination and autophagy processes. Moreover, PRC1, TOP2A and CKAP2L may be the hub genes involved in HCC tumorigenesis, and their biomarker roles were further demonstrated via Gene Expression Profiling Interactive Analysis (GEPIA) and Oncomine databases. In addition, the levels of PRC1, TOP2A and CKAP2L were obviously up-regulated in the sera of HCC patients. Conclusion PRC1, TOP2A and CKAP2L may serve as biomarkers for the prognostic prediction of HCC patients.
Collapse
Affiliation(s)
- Junting Huang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Yating Zhan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Lili Jiang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Yuxiang Gao
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Binyu Zhao
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Yuxiao Zhang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Wenjie Zhang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Jianjian Zheng
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Jinglu Yu
- Department of Laboratory Medicine, Lishui Municipal Central Hospital, Lishui, People's Republic of China.,The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, People's Republic of China
| |
Collapse
|
13
|
Zhang Q, Jiang J. Regulation of Hedgehog Signal Transduction by Ubiquitination and Deubiquitination. Int J Mol Sci 2021; 22:ijms222413338. [PMID: 34948134 PMCID: PMC8703657 DOI: 10.3390/ijms222413338] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 12/23/2022] Open
Abstract
The Hedgehog (Hh) family of secreted proteins governs embryonic development and adult tissue homeostasis in species ranging from insects to mammals. Deregulation of Hh pathway activity has been implicated in a wide range of human disorders, including congenital diseases and cancer. Hh exerts its biological influence through a conserved signaling pathway. Binding of Hh to its receptor Patched (Ptc), a twelve-span transmembrane protein, leads to activation of an atypical GPCR family protein and Hh signal transducer Smoothened (Smo), which then signals downstream to activate the latent Cubitus interruptus (Ci)/Gli family of transcription factors. Hh signal transduction is regulated by ubiquitination and deubiquitination at multiple steps along the pathway including regulation of Ptc, Smo and Ci/Gli proteins. Here we review the effect of ubiquitination and deubiquitination on the function of individual Hh pathway components, the E3 ubiquitin ligases and deubiquitinases involved, how ubiquitination and deubiquitination are regulated, and whether the underlying mechanisms are conserved from Drosophila to mammals.
Collapse
Affiliation(s)
- Qing Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing 210061, China
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing 210061, China
- Correspondence: (Q.Z.); (J.J.)
| | - Jin Jiang
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence: (Q.Z.); (J.J.)
| |
Collapse
|
14
|
Immunomodulation: An immune regulatory mechanism in carcinoma therapeutics. Int Immunopharmacol 2021; 99:107984. [PMID: 34303999 DOI: 10.1016/j.intimp.2021.107984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/29/2021] [Accepted: 07/11/2021] [Indexed: 01/01/2023]
Abstract
Cancer has been generally related to the possession of numerous mutations which interrupt important signaling pathways. Nevertheless, deregulated immunological signaling is considered as one of the key factors associated with the development and progression of cancer. The signaling pathways operate as modular network with different components interacting in a switch-like fashion with two proteins interplaying between each other leading to direct or indirect inhibition or stimulation of down-stream factors. Genetic, epigenetic, and transcriptomic alterations maintain the pathological conduit of different signaling pathways via affecting diverse mechanisms including cell destiny. At present, immunotherapy is one of the best therapies opted for cancer treatment. The cancer immunotherapy strategy includes harnessing the specificity and killing mechanisms of the immunological system to target and eradicate malignant cells. Targeted therapies utilizing several little molecules including Galunisertib, Astragaloside-IV, Melatonin, and Jervine capable of regulating key signaling pathways can effectively help in the management of different carcinomas.
Collapse
|
15
|
Chen J, Wen B, Wang Y, Wu S, Zhang X, Gu Y, Wang Z, Wang J, Zhang W, Yong J. Jervine exhibits anticancer effects on nasopharyngeal carcinoma through promoting autophagic apoptosis via the blockage of Hedgehog signaling. Biomed Pharmacother 2020; 132:110898. [PMID: 33113432 DOI: 10.1016/j.biopha.2020.110898] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 12/30/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant tumor originating from the superior mucosal epithelium of the nasopharynx. However, effective therapies for NPC are still required. Reducing Hedgehog signaling pathway has been shown to suppress tumor growth. In this study, we attempted to explore whether Jervine (JV), an inhibitor of Hedgehog signaling, had anti-cancer effects on NPC, and the underlying mechanisms. Our findings showed that JV treatments markedly reduced the proliferation of NPC cells in a dose- and time-dependent manner. Cell cycle arrest in G2/M phase was significantly enhanced by JV, along with evident DNA damage. Moreover, JV treatment effectively induced apoptosis in NPC cells through improving Caspase-3 activation. Furthermore, ROS production and mitochondrial impairments were detected in JV-incubated NPC cells with elevated releases of Cyto-c from mitochondria. JV also dramatically triggered autophagy through blocking AKT/mTOR and increasing AMPK signaling pathways. Intriguingly, we showed that JV-induced apoptosis was mainly via an autophagy-dependent manner. In addition, the expression levels of SHH, PTCH1, SMO and GLI1 were markedly suppressed in NPC cells, demonstrating the hindered Hedgehog signaling. Importantly, we found that JV-induced apoptosis and autophagy were closely associated with the blockage of Hedgehog signaling. Our in vivo studies confirmed the anti-cancer effects of JV on NPC through inducing autophagy, as evidenced by the markedly reduced tumor growth rate and weight without side effects and toxicity. Taken together, JV may be a promising and effective agent for human NPC treatment through repressing Hedgehog signaling pathway and inducing autophagic cell death.
Collapse
Affiliation(s)
- Jing Chen
- Department of Pathology, Jingjiang People's Hospital, Jingjiang, Jiangsu, 214500, China
| | - Bin Wen
- Department of Oncology, Jingjiang Chinese Medicine Hospital, Jingjiang, Jiangsu, 214500, China
| | - Yu Wang
- Department of Pathology, Jingjiang People's Hospital, Jingjiang, Jiangsu, 214500, China
| | - Sheng Wu
- Department of Pathology, Jingjiang People's Hospital, Jingjiang, Jiangsu, 214500, China
| | - Xuesong Zhang
- Central Laboratory, Jingjiang People's Hospital, Jingjiang, Jiangsu, 214500, China
| | - Yonggui Gu
- Department of Otolaryngology, Jingjiang People's Hospital, Jingjiang, Jiangsu, 214500, China
| | - Zhiyi Wang
- Department of Otolaryngology, East Theater General Hospital of PLA, Nanjing, Jiangsu, 210000, China
| | - Jianjiang Wang
- Department of General Surgery, Jingjiang People's Hospital, Jingjiang, Jiangsu, 214500, China
| | - Wenzhong Zhang
- Department of Otolaryngology, East Theater General Hospital of PLA, Nanjing, Jiangsu, 210000, China
| | - Ji Yong
- Department of Otolaryngology, East Theater General Hospital of PLA, Nanjing, Jiangsu, 210000, China.
| |
Collapse
|
16
|
Qiu J, Zhou S, Cheng W, Luo C. LINC00294 induced by GRP78 promotes cervical cancer development by promoting cell cycle transition. Oncol Lett 2020; 20:262. [PMID: 32989396 PMCID: PMC7517597 DOI: 10.3892/ol.2020.12125] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 05/27/2020] [Indexed: 12/17/2022] Open
Abstract
Cervical cancer is one of the most common gynecological malignancies, and it has become a crucial public health problem. In the present study, the expression profiles of cervical cancer and normal cervical tissues were downloaded from the Gene Expression Omnibus and The Cancer Genome Atlas databases. Subsequently, the dysregulated long non-coding RNAs (lncRNAs) in cervical cancer were identified using R software Differentially expressed lncRNAs in cervical cancer that were associated with glucose-regulated protein 78 (GRP78) were screened out and the results demonstrated that eight lncRNAs were strongly positively correlated with GRP78. In order to confirm the relationship between GRP78 and candidate lncRNAs, GRP78 small interfering RNA (siRNA) was transfected into HeLa cells. The target lncRNAs that were regulated by GRP78 were then identified by reverse transcription-quantitative PCR and it was revealed that LINC00294 was significantly downregulated following GRP78-knockdown. Subsequently, Gene Set Enrichment Analysis demonstrated that LINC00294 was mainly enriched in regulating the cell cycle and the Hedgehog pathway. Following transfection of HeLa and SiHa cells with LINC00294 siRNA, the cell cycle was arrested at the G0/G1 phase. Western blotting suggested that LINC00294-knockdown downregulated the expression of cell cycle-associated factors (cyclin D, cyclin E and cyclin Dependent kinase 4) and upregulated cell cycle inhibitory factors (p16 and p21). The Hedgehog pathway was inhibited following knockdown of LINC00294 in HeLa and SiHa cells. In summary, LINC00294 induced by GRP78 promoted the progression of cervical cancer by regulating the cell cycle via Hedgehog pathway.
Collapse
Affiliation(s)
- Jiangnan Qiu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Shulin Zhou
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Wenjun Cheng
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Chengyan Luo
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| |
Collapse
|