1
|
Zhu Y, Chen Z, Niu K, Li M, Deng Y, Zhang J, Wei D, Wang J, Zhao Y. USP33 Regulates DNA Damage Response and Carcinogenesis Through Deubiquitylating and Stabilising p53. Cell Prolif 2025; 58:e13793. [PMID: 39694539 PMCID: PMC12099211 DOI: 10.1111/cpr.13793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
The de-ubiquitinase USP33 has been shown to possess either tumour-promoting or inhibitory effect on human cancer cells. However, all these findings are mainly based on in vitro cell culture models, and the in vivo evidence, which is more plausible to digest the functional role of USP33 in carcinogenic process, is still lacking. Here, we demonstrate that USP33 modulates DNA damage responses including cell cycle arrest and apoptosis induction through associating with p53. It directly interacts with p53 to mediate its de-ubiquitination and further stabilisation under DNA damage condition. Depletion of USP33 induces an enhanced level of p53 ubiquitination, which de-stabilises p53 protein leading to impaired DNA damage responses. Furthermore, USP33 silencing shows either promoted or inhibited effect on cell proliferation in human cancer cells with p53 WT and mutant background, respectively. Consistently, mice with hepatocyte-specific USP33 knockout are more sensitive to nitrosodiethylamine (DEN)-induced hepatocarcinogenesis compared to wild type mice. Thus, our in vitro and in vivo evidences illustrate that USP33 possesses anti-tumour activity via regulating p53 stability and activity.
Collapse
Affiliation(s)
- Yuqi Zhu
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Zixiang Chen
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
| | - Kaifeng Niu
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
| | - Mengge Li
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yuchun Deng
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Ji Zhang
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Di Wei
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
| | - Jiaqi Wang
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - YongLiang Zhao
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
2
|
Hang Q, Zuo S, Yang Y, Wang Y, Li C, Li W, Guo J, Hou S, Huang H. USP33 is an integrin α6 deubiquitinase and promotes esophageal squamous cell carcinoma cell migration and metastasis. J Cancer Res Clin Oncol 2024; 150:511. [PMID: 39589547 PMCID: PMC11599434 DOI: 10.1007/s00432-024-06041-5] [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: 10/22/2024] [Accepted: 11/16/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE The deubiquitinating enzymes (DUBs) have been linked to cancer initiation and progression. Although ubiquitin-specific protease 33 (USP33) represents a significant factor in regulating various tumor cell behaviors, its specific biological functions and precise mechanisms in esophageal squamous cell carcinoma (ESCC) progression remain unclear. METHODS The expressions of USP33 mRNA in GEO databases, clinical ESCC samples, and USP33 protein were analyzed using bioinformatics, RT-PCR, and immunohistochemistry, respectively. Using Kaplan-Meier survival curves, the log-rank test was used to determine the cumulative survival rate. Western blotting was used to determine indicated protein expression. The cell biological functions were evaluated by cell growth assay, transwell, cell adhesion, and cell spreading assay, respectively. The interaction between USP33 and integrins was detected by immunoprecipitation, and the deubiquitination was performed by deubiquitination assay. The metastatic ability was checked by tail vein injection. RESULTS A significant positive correlation was found between USP33 expression and clinical TNM stage, T classification, and poor prognosis in patients with ESCC. USP33 promoted laminin-dependent adhesion, spreading, and migration of ESCC cells but not their proliferation. Mechanistically, USP33 mediates cell migration through binding, deubiquinating, and stabilizing integrin α6. USP33 knockdown could inhibit ESCC cell migration and metastasis majorly through integrin α6. CONCLUSION This study reveals a novel mechanism of USP33 in promoting laminin-dependent ESCC cell migration and metastasis through integrin α6, suggesting that USP33 may be a promising target for treating ESCC.
Collapse
Affiliation(s)
- Qinglei Hang
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China.
- Jiangsu Provincial Innovation and Practice Base for Postdoctors, Suining People's Hospital, Affiliated Hospital of Xuzhou Medical University, Suining, Jiangsu Province, 221200, China.
| | - Shiying Zuo
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Yawen Yang
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
| | - Yuanzhi Wang
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Caimin Li
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Wenqian Li
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Jingya Guo
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Sicong Hou
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China.
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China.
| | - Haifeng Huang
- Department of Laboratory Medicine, The First People's Hospital of Yancheng, Yancheng, Jiangsu Province, 224006, China.
- Department of Laboratory Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224006, China.
| |
Collapse
|
3
|
Zhou Y, Liao Y, Fan L, Wei X, Huang Q, Yang C, Feng W, Wu Y, Gao X, Shen X, Zhou J, Xia Z, Zhang Z. Lung-Targeted Lipid Nanoparticle-Delivered siUSP33 Attenuates SARS-CoV-2 Replication and Virulence by Promoting Envelope Degradation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406211. [PMID: 39301916 PMCID: PMC11558077 DOI: 10.1002/advs.202406211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/02/2024] [Indexed: 09/22/2024]
Abstract
As a structural protein of SARS-CoV-2, the envelope (E) protein not only plays a key role in the formation of viral particles, but also forms ion channels and has pathogenic functions, including triggering cell death and inflammatory responses. The stability of E proteins is controlled by the host ubiquitin-proteasome system. By screening human deubiquitinases, it is found that ubiquitin-specific protease 33 (USP33) can enhance the stability of E proteins depending on its deubiquitinase activity, thereby promoting viral replication. In the absence of USP33, E proteins are rapidly degraded, leading to a reduced viral load and inflammation. Using lipid nanoparticle (LNP) encapsulation of siUSP33 by adjusting the lipid components (ionizable cationic lipids), siUSP33 is successfully delivered to mouse lung tissues, rapidly reducing USP33 expression in the lungs and maintaining knockdown for at least 14 days, effectively suppressing viral replication and virulence. This method of delivery allows efficient targeting of the lungs and a response to acute infections without long-term USP33 deficiency. This research, based on the deubiquitination mechanism of USP33 on the E protein, demonstrates that LNP-mediated siRNA delivery targeting USP33 plays a role in antiviral and anti-inflammatory responses, offering a novel strategy for the prevention and treatment of SARS-CoV-2.
Collapse
Affiliation(s)
- Yuzheng Zhou
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Yujie Liao
- Department of Cell BiologySchool of Life SciencesCentral South UniversityChangsha410083China
| | - Lujie Fan
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
- Guangzhou LaboratoryGuangzhou510700China
| | - Xiafei Wei
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Qiang Huang
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Chuwei Yang
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Wei Feng
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Yezi Wu
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Xiang Gao
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Xiaotong Shen
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Jian Zhou
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
| | - Zanxian Xia
- Department of Cell BiologySchool of Life SciencesCentral South UniversityChangsha410083China
- Hunan Key Laboratory of Animal Models for Human DiseasesHunan Key Laboratory of Medical Genetics & Center for Medical GeneticsSchool of Life SciencesCentral South UniversityChangsha410013China
| | - Zheng Zhang
- Institute for HepatologyNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalThe Second Affiliated HospitalSchool of MedicineSouthern University of Science and TechnologyShenzhen518112China
- Shenzhen Research Center for Communicable Disease Diagnosis and TreatmentChinese Academy of Medical SciencesShenzhen518112China
| |
Collapse
|
4
|
Tanna B, Yadav S, Patel MK, Mishra A. Metabolite Profiling, Biological and Molecular Analyses Validate the Nutraceutical Potential of Green Seaweed Acrosiphoniaorientalis for Human Health. Nutrients 2024; 16:1222. [PMID: 38674913 PMCID: PMC11055090 DOI: 10.3390/nu16081222] [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: 03/15/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Seaweeds have proven to be nutrient-dense and are rich in antioxidants, like phenolics, flavonoids, and other essential metabolites that help to provide their medicinal benefits. Non-targeted metabolite profiling of the tropical green seaweed Acrosiphonia orientalis showed the presence of numerous groups of contents, including sugars, essential amino acids, and fatty acids. Targeted metabolite profiling using HPLC identified 17 amino acids. The extract exhibited a very low half-maximal effective concentration (EC50) dosage for HeLa and Huh-7 cell lines, indicating a high likelihood of anticancer properties. A significant positive correlation was found between biological activities, such as antioxidation, scavenging, and reducing power with the phenolic and flavonoid contents. The extract revealed augmentation of proliferation in selected cervical cells, as it upregulated p53 1.3-fold, and downregulated important cancerous genes such as Cas-3 and DNMT 12- and 8-fold, respectively. An approximate 55-fold downregulation was observed in selected hepatic cell lines. Microarray analysis of hepatic cells indicated 0.27% and 0.07% upregulation of coding and non-coding genes, respectively, and 0.41% and 0.13% downregulation of coding and non-coding genes, respectively. As a consequence, it can be said that A. orientalis has possible medicinal use, such as anticancer activity, and therefore may be an intriguing food component that has potential as a regular dietary supplement.
Collapse
Affiliation(s)
- Bhakti Tanna
- Division of Biotechnology and Phycology, CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar 364002, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sonam Yadav
- Division of Biotechnology and Phycology, CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar 364002, India
| | - Manish Kumar Patel
- Division of Biotechnology and Phycology, CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar 364002, India
- Department of Postharvest Science of Fresh Produce, Agricultural Research Organization (ARO), Volcani Center, Rishon LeZion 7505101, Israel
| | - Avinash Mishra
- Division of Biotechnology and Phycology, CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar 364002, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
5
|
Feng L, Shu HP, Sun LL, Tu YC, Liao QQ, Yao LJ. Role of the SLIT-ROBO signaling pathway in renal pathophysiology and various renal diseases. Front Physiol 2023; 14:1226341. [PMID: 37497439 PMCID: PMC10366692 DOI: 10.3389/fphys.2023.1226341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 06/30/2023] [Indexed: 07/28/2023] Open
Abstract
SLIT ligand and its receptor ROBO were initially recognized for their role in axon guidance in central nervous system development. In recent years, as research has advanced, the role of the SLIT-ROBO signaling pathway has gradually expanded from axonal repulsion to cell migration, tumor development, angiogenesis, and bone metabolism. As a secreted protein, SLIT regulates various pathophysiological processes in the kidney, such as proinflammatory responses and fibrosis progression. Many studies have shown that SLIT-ROBO is extensively involved in various aspects of kidney development and maintenance of structure and function. The SLIT-ROBO signaling pathway also plays an important role in different types of kidney disease. This article reviews the advances in the study of the SLIT-ROBO pathway in various renal pathophysiological and kidney disorders and proposes new directions for further research in this field.
Collapse
|
6
|
Liu X, Xu J, Shen B, Xu J, Jiang J. USP33 promotes pancreatic cancer malignant phenotype through the regulation of TGFBR2/TGFβ signaling pathway. Cell Death Dis 2023; 14:362. [PMID: 37322017 PMCID: PMC10272277 DOI: 10.1038/s41419-023-05871-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 04/07/2023] [Accepted: 05/31/2023] [Indexed: 06/17/2023]
Abstract
Pancreatic cancer (PC) ranked fourth among cancer-related death worldwide with a survival rate less than 5%. The abnormal proliferation and distant metastasis are major obstacles for the diagnosis and treatment of pancreatic cancer, therefore, it is urgent for researchers to uncover the molecular mechanisms underlying the PC proliferation and metastasis. In current study, we found that USP33, a member of deubiquitinating enzyme family, was upregulated among PC samples and cells, meanwhile, the high expression of USP33 correlated with poor prognosis of patients. Function experiments revealed that USP33 overexpression promoted the proliferation, migration and invasion of PC cells while the inhibition of USP33 expression in PC cells exhibited the opposite effect. The mass spectrum and luciferase complementation assay screened TGFBR2 as the potential binding protein of USP33. Mechanistically, USP33 triggered the deubiquitination of TGFBR2 and prevented its degradation by lysosome, therefore promoted TGFBR2 accumulation in cell membrane and eventually contributed to the sustained activation of TGF-β signaling. Moreover, our results revealed that the activation of TGF-β targeted gene ZEB1 promoted the transcription of USP33. In conclusion, our study found that USP33 contributed to the proliferation and metastasis of pancreatic cancer through a positive feedback loop with TGF-β signaling pathway. Moreover, this study suggested that USP33 may serve as a potential prognostic and therapeutic target in PC.
Collapse
Affiliation(s)
- Xinyuan Liu
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jian Xu
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bingbing Shen
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jichuan Xu
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jianxin Jiang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
7
|
Han PP, Zhang GQ, Li L, Yue L. Downregulation of USP33 inhibits Slit/Robo signaling pathway and is associated with poor patient survival of glioma. J Neurosurg Sci 2023; 67:113-120. [PMID: 32972109 DOI: 10.23736/s0390-5616.20.04929-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Gliomas are the most common malignant tumors in the central nervous system originating from brain glial cells. Although characterized as highly invasive and highly malignant, few molecular targeting therapies have been developed. Ubiquitin Specific Protease 33 (USP33), a gene encoding a deubiquinating enzyme important in a variety of processes, including Slit-dependent cell migration and beta-2 adrenergic receptor signaling, participates in the development of several malignant tumors, however, its role in the development of glioma has not been evaluated. METHODS Real-time quantitative PCR was performed to examine the expression of USP33 in glioma tissues and cell lines. Immunohistochemistry was performed to determine USP33 expressions in glioma tissue microarray. Transwell assay was performed to analyze the effect of USP33 on glioma cell line migration. The Kaplan-Meier method and log rank test were applied to evaluate the prognostic value of USP33 expression. Univariate and multivariate Cox proportional hazard regression models were used to identify the independent prognostic factors associated with overall survival (OS) or disease-free survival. RESULTS The present study demonstrated that USP33 expression was significantly downregulated in glioma tissues. Lower expression of USP33 was associated with a poorer patient disease-free survival and overall survival. In vitro studies revealed that overexpression of USP33 significantly inhibited the migration ability of glioma cells. Mechanistically, USP33 inhibits glioma cell migration by regulating the function of Slit/Robo signaling pathway. CONCLUSIONS Downregulation of USP33 is associated with poor patient survival of glioma. USP33 inhibits glioma cell migration by Slit/Robo signaling pathway. This mechanism may be applied for development of targeting therapy especially for the high-grade glioma.
Collapse
Affiliation(s)
- Peng-Peng Han
- Second Department of Neurology, Weifang Yidu Central Hospital, Weifang, Shandong, China
| | - Gui-Qin Zhang
- Second Department of Neurology, Weifang Yidu Central Hospital, Weifang, Shandong, China
| | - Lin Li
- Department of Gynecology Ward II, Weifang Yidu Central Hospital, Weifang, China
| | - Liang Yue
- Department of Surgery Teaching and Research Section, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Haike Mingcheng, Chengdu, China -
| |
Collapse
|
8
|
USPs in Pancreatic Ductal Adenocarcinoma: A Comprehensive Bioinformatic Analysis of Expression, Prognostic Significance, and Immune Infiltration. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6109052. [PMID: 36582601 PMCID: PMC9794441 DOI: 10.1155/2022/6109052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/12/2022] [Accepted: 11/16/2022] [Indexed: 12/24/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), as an intractable malignancy, still causes an extremely high mortality worldwide. The ubiquitin-specific protease (USP) family constitutes the major part of deubiquitinating enzymes (DUBs) which has been reported to be involved in initiation and progression of various malignancies via the function of deubiquitination. However, the biological function and clinical values of USPs in PDAC have not been comprehensively elucidated. In this study, Gene Expression Profiling Interactive Analysis (GEPIA), Gene Expression Omnibus (GEO) datasets, UALCAN database, and the Human Protein Atlas (HPA) online tool were used to analyze the expression level and the relationship between USP expression and clinicopathological features in PDAC. Survival module of HPA and Kaplan-Meier plotter (KMP) databases was recruited to explore the prognostic value of USPs. Tumor Immune Estimation Resource (TIMER) online tool and KMP databases were utilized to elucidate tumor immune infiltration and immune-related survival of USPs. CBioPortal online tool was used to identify the gene mutation level of USPs in PDAC. Both cBioPortal and LinkedOmics were used to confirm the potential biological functions of USPs in PDAC. Our study showed that USP10, USP14, USP18, USP32, USP33, and USP39 (termed as six-USPs) expressions were significantly elevated in tumor tissues. The high expression of the four USPs (USP10, USP14, USP18, and USP39) indicated a poor prognosis. A significant relationship was indicated between the expression of six-USPs and clinicopathological features. Also, the expression of six-USPs was related to promoter methylation level. Moreover, more than 40% genetic alterations and mutations were discovered in six-USPs. Furthermore, the six-USP expression was correlated with immune infiltration and immune-related prognosis. The functional analysis found that the six-USPs were involved in various biological processes and signaling pathways, such as nucleocytoplasmic transport, choline metabolism in cancer, cell cycle, ErbB signaling pathway, RIG-I-like receptor signaling pathway, TGF-β signaling pathway, and TNF signaling pathway. In conclusion, the results showed that six-USPs are potential prognostic biomarkers and can be recruited as possible therapeutic targets of PDAC.
Collapse
|
9
|
Hu J, Chen W, Shen L, Chen Z, Huang J. Crosstalk between the peripheral nervous system and breast cancer influences tumor progression. Biochim Biophys Acta Rev Cancer 2022; 1877:188828. [PMID: 36283598 DOI: 10.1016/j.bbcan.2022.188828] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/18/2022]
Abstract
Recent studies have shown that peripheral nerves play an important role in the progression of breast cancer. Breast cancer cells (BCCs) promote local peripheral nerve growth and branching by secreting neuroactive molecules, including neurotrophins and axon guidance molecules (AGMs). Sympathetic nerves promote breast cancer progression, while parasympathetic and sensory nerves mainly have anti-tumor effects in the progression of breast cancer. Specifically, peripheral nerves can influence the progression of breast cancer by secreting neurotransmitters not only directly binding to the corresponding receptors of BCCs, but also indirectly acting on immune cells to modulate anti-tumor immunity. In this review, we summarize the crosstalk between breast cancer and peripheral nerves and the roles of important neuroactive molecules in the progression of breast cancer. In addition, we summarize indicators, including nerve fiber density and perineural invasion (PNI), that may help determine the prognosis of breast cancer based on current research results, as well as potential therapeutic approaches, such as β-blockers and retroviral-mediated genetic neuroengineering techniques, that may enhance the prognosis of breast cancer. In addition, we propose suggestions for future research priorities based on a current lack of knowledge in this area.
Collapse
Affiliation(s)
- Jianming Hu
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Wuzhen Chen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lesang Shen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Zhigang Chen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China..
| | - Jian Huang
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China..
| |
Collapse
|
10
|
Gu H, Li Y, Cui X, Cao H, Hou Z, Ti Y, Liu D, Gao J, Wang Y, Wen P. MICAL1 inhibits colorectal cancer cell migration and proliferation by regulating the EGR1/β-catenin signaling pathway. Biochem Pharmacol 2022; 195:114870. [PMID: 34902339 DOI: 10.1016/j.bcp.2021.114870] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/18/2021] [Accepted: 12/03/2021] [Indexed: 12/26/2022]
Abstract
MICAL1 has been reported to be involved in the malignant processes of several types of cancer cells, however, the roles of MICAL1 in colorectal cancer (CRC) have not been well-characterized. This study aims to investigate the cellular functions and molecular mechanisms of MICAL1 in CRC cells. Here, we found that both mRNA and protein levels of MICAL1 were down-regulated in colorectal cancer tissues compared with matched adjacent non-tumor tissues, and the expression level of MICAL1 was correlated with the metastatic status of colorectal cancer. Importantly, overexpression of MICAL1 significantly inhibited colorectal cancer cell migration and growth, and increased the level of E-cadherin and Occludin, and suppressed the expression level of Vimentin and N-cadherin; while silencing of MICAL1 promoted CRC cell migration and enhanced EMT. In addition, MICAL1 overexpression significantly inhibited the proliferation and growth of CRC in vitro and in vivo. Moreover, RNA sequencing and bioinformatics analysis identified that MICAL1 was closely correlated with "cell migration", "cell cycle" and "β-catenin signaling" genesets. Mechanistically, overexpression of MICAL1 downregulated the mRNA level of EGR1 and β-catenin, decreased the protein level and nuclear translocation of β-catenin, and inhibited the transcriptions of β-catenin downstream targets, c-myc and cyclin D1. The ectopic expression of EGR1 or β-catenin can significantly block the MICAL1-mediated inhibitory effects. Collectively, MICAL1 is down-regulated in CRC, and plays an inhibitory role in the migration and growth of CRC cells by suppressing the ERG1/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Huanyu Gu
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Yi Li
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Xiuping Cui
- Life Science Institute, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Huiru Cao
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Zhijuan Hou
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Yunhe Ti
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Dahua Liu
- Biological Anthropology Institute, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Jing Gao
- Department of Ultrasonography, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Yu Wang
- Life Science Institute, Jinzhou Medical University, Jinzhou 121000, Liaoning, China.
| | - Pushuai Wen
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121000, Liaoning, China; Biological Anthropology Institute, Jinzhou Medical University, Jinzhou 121000, Liaoning, China.
| |
Collapse
|
11
|
Wang H, Liu Z, Sun Z, Zhou D, Mao H, Deng G. Ubiquitin specific peptidase 33 promotes cell proliferation and reduces apoptosis through regulation of the SP1/PI3K/AKT pathway in retinoblastoma. Cell Cycle 2021; 20:2066-2076. [PMID: 34470581 DOI: 10.1080/15384101.2021.1970305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Ubiquitin-specific protease 33 (USP33), a deubiquitinating enzyme (DUB), has been identified to serve as a tumor suppressor or an oncogene in different cancers. However, its role in retinoblastoma (RB) remains unknown. Here, we aimed to uncover USP33 expression profile and function in RB, and disclose the underlying mechanism. USP33 levels in RB tissues and cells were determined using RT-qPCR and western blotting assays. USP33 effects on cell growth, cycle, apoptosis and tumorigenesis were studied using MTT, Edu, cycle and western blotting and in vivo assays. The results showed that USP33 expression levels were elevated in RB tissues and cells as compared with normal retinal tissues and cells. Downregulation of USP33 in RB Y79 and WERI-RB1 cells leaded to significant increases in cell apoptosis, G1 phase arrest and tumorigenesis, and reductions in cell growth and G2 and S phase arrest, as well as inhibited the activation of the PI3K/AKT signaling. SP1 overexpression abolished the roles of USP33 downregulation in modulating the activation of PI3K/AKT signaling, cell growth, apoptosis, and cell cycle. This study uncovered that USP33 promoted the progression of RB through regulation of the SP1/PI3K/AKT pathway.
Collapse
Affiliation(s)
- Hao Wang
- Department of Ophthalmology, The Third People's Hospital of Changzhou, Changzhou City, Jiangsu Province, China
| | - Zhinan Liu
- Department of Ophthalmology, The Third People's Hospital of Changzhou, Changzhou City, Jiangsu Province, China
| | - Zhuo Sun
- Department of Ophthalmology, The Third People's Hospital of Changzhou, Changzhou City, Jiangsu Province, China
| | - Dong Zhou
- Department of Ophthalmology, The Third People's Hospital of Changzhou, Changzhou City, Jiangsu Province, China
| | - Hanyan Mao
- Department of Ophthalmology, The Third People's Hospital of Changzhou, Changzhou City, Jiangsu Province, China
| | - Guohua Deng
- Department of Ophthalmology, The Third People's Hospital of Changzhou, Changzhou City, Jiangsu Province, China
| |
Collapse
|
12
|
Ding C, Li Y, Wang S, Xing C, Chen L, Zhang H, Wang Y, Dai M. ROBO2 hampers malignant biological behavior and predicts a better prognosis in pancreatic adenocarcinoma. Scand J Gastroenterol 2021; 56:955-964. [PMID: 34148491 DOI: 10.1080/00365521.2021.1930144] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a fatalmalignant cancer with extremely poor prognosis and high mortality. Genome wide studies show that Slit/Robo signaling pathway takes a major effect in the oncogenesis and progression of pancreatic cancer. However, the function and mechanism of ROBO2 in the development of PDAC remains unclear. METHODS In present study, we use Western blot and real-time polymerase chain reaction (RT-PCR) to detect the expression of ROBO2 in pancreatic cell lines. Cell proliferation,Transwellmigration and invasion were conducted inAsPC-1, MIA PaCa-2 and PANC-1cell lines. RNA sequencing, bioinformatics analysisand Western blot were used to explore its mechanism and potential target molecules. The expression of ROBO2 in 95 tumor tissues was detected by immunohistochemistry. RESULTS ROBO2 expression was downregulated in PDAC cell lines and tissue samples. A high expression of ROBO2 was associated with better prognosis. Upregulation of ROBO2 inhibited PDAC cell proliferation, migration, and invasion. However, we found theoppositeresults in the ROBO2 downregulation group. In addition, the function of ROBO2 on cell proliferation was further affirmed by the animal model. Finally, the results of RNA sequencing indicated that ROBO2 partly promoted the antitumor activity by inhibiting ECM1 in PDAC. CONCLUSIONS Our work suggests that ROBO2 inhibits tumor progression in PDAC and may serve as a predictive biomarker and therapeutic target in PDAC.
Collapse
Affiliation(s)
- Cheng Ding
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yatong Li
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Shunda Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Cheng Xing
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lixin Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Hanyu Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yizhi Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Menghua Dai
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
13
|
Ding T, Zhu Y, Jin H, Zhang P, Guo J, Zheng J. Circular RNA circ_0057558 Controls Prostate Cancer Cell Proliferation Through Regulating miR-206/USP33/c-Myc Axis. Front Cell Dev Biol 2021; 9:644397. [PMID: 33718387 PMCID: PMC7952531 DOI: 10.3389/fcell.2021.644397] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 01/26/2021] [Indexed: 12/25/2022] Open
Abstract
We previously reported the elevated expression of circ_0057558 in prostate cancer tissues and cell lines. Here, we aimed to determine the biological function of circ_0057558 in prostate cancer. In the current study, circ_0057558 knockdown in prostate cancer cells significantly repressed cell proliferation and colony formation, but promoted cell arrest and enhanced the sensitivity to docetaxel. Bioinformatics analysis prediction and RNA-pull down assay identified miR-206 as the potential binding miRNA of circ_0057558. A negative correlation was observed between the expression of miR-206 and circ_0057558 in prostate cancer tissues. miR-206 mimics rescued the function of circ_0057558 overexpression on prostate cancer cells. Further, the bioinformatics analysis and luciferase assay suggested that miR-206 may target ubiquitin-specific peptidase 33 (USP33). USP33 mRNA expression has negative correlation with miR-206 expression and positive correlation with circ_0057558 expression in prostate cancer tissues. USP33 overexpression partially blocked the effects of miR-206 mimics on prostate cell proliferation. USP33 could bind and deubiquitinate c-Myc. Increased c-Myc protein by circ_0057558 overexpression was partially reversed by miR-206 mimics. The proliferation inhibition activity of MYC inhibitor 361 (MYCi361) was more prominent in primary prostate cancer cells and patient-derived xenograft (PDX) model with higher level of circ_0057558. Collectively, circ_0057558 gives an impetus to cell proliferation and cell cycle control in prostate cancer cell lines by sponging miR-206 and positively regulating the transcription of the miR-206 target gene USP33.
Collapse
Affiliation(s)
- Tao Ding
- Department of Urology, The Sixth People's Hospital South Campus, Shanghai, China
| | - Yanjun Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huimin Jin
- Department of Laboratory Medicine, Zhoupu Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Ping Zhang
- Department of Laboratory Medicine, Zhoupu Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianghua Zheng
- Department of Laboratory Medicine, Zhoupu Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
14
|
Ye DX, Wang SS, Huang Y, Wang XJ, Chi P. USP43 directly regulates ZEB1 protein, mediating proliferation and metastasis of colorectal cancer. J Cancer 2021; 12:404-416. [PMID: 33391437 PMCID: PMC7738986 DOI: 10.7150/jca.48056] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/06/2020] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer is one of the most common malignant tumors of the digestive tract. In this study, we had examined the biological role of USP43 in colorectal cancer. USP43 protein and mRNA abundance in clinical tissues and five cell lines were analyzed with quantitative real-time PCR test (qRT-PCR) and western blot. USP43 overexpression treated DLD1 cells and USP43 knockdown treated SW480 cells were used to study cell proliferation, migration, colony formation, invasion, and the expression of epithelial-mesenchymal transformation (EMT) biomarkers. Moreover, ubiquitination related ZEB1 degradation was studied with qRT-PCR and western blot. The relationships between USP43 and ZEB1 were investigated with western blot, co-immunoprecipitation, migration, and invasion. USP43 was highly expressed in colorectal cancer tissues. USP43 overexpression and knockdown treatments could affect cell proliferation, colony formation, migration, invasion, and the expression of EMT associated biomarkers. Moreover, USP43 can regulate ZEB1 degradation through ubiquitination pathway. USP43 could promote the proliferation, migration, and invasion of colorectal cancer. Meanwhile, USP43 can deubiquitinate and stabilize the ZEB1 protein, which plays an important role in the function of colorectal cancer.
Collapse
Affiliation(s)
- Dao-Xiong Ye
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Si-Si Wang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Ying Huang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Xiao-Jie Wang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Pan Chi
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| |
Collapse
|
15
|
Sengupta D, Bhattacharya G, Ganguli S, Sengupta M. Structural insights and evaluation of the potential impact of missense variants on the interactions of SLIT2 with ROBO1/4 in cancer progression. Sci Rep 2020; 10:21909. [PMID: 33318575 PMCID: PMC7736846 DOI: 10.1038/s41598-020-78882-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023] Open
Abstract
The cognate interaction of ROBO1/4 with its ligand SLIT2 is known to be involved in lung cancer progression. However, the precise role of genetic variants, disrupting the molecular interactions is less understood. All cancer-associated missense variants of ROBO1/4 and SLIT2 from COSMIC were screened for their pathogenicity. Homology modelling was done in Modeller 9.17, followed by molecular simulation in GROMACS. Rigid docking was performed for the cognate partners in PatchDock with refinement in HADDOCK server. Post-docking alterations in conformational, stoichiometric, as well as structural parameters, were assessed. The disruptive variants were ranked using a weighted scoring scheme. In silico prioritisation of 825 variants revealed 379 to be potentially pathogenic out of which, about 12% of the variants, i.e. ROBO1 (14), ROBO4 (8), and SLIT2 (23) altered the cognate docking. Six variants of ROBO1 and 5 variants of ROBO4 were identified as "high disruptors" of interactions with SLIT2 wild type. Likewise, 17 and 13 variants of SLIT2 were found to be "high disruptors" of its interaction with ROBO1 and ROBO4, respectively. Our study is the first report on the impact of cancer-associated missense variants on ROBO1/4 and SLIT2 interactions that might be the drivers of lung cancer progression.
Collapse
Affiliation(s)
- Debmalya Sengupta
- Department of Genetics, University of Calcutta, University College of Science (UCSTA), 35, Ballygunge Circular Road, Kolkata, 700 019, India
| | - Gairika Bhattacharya
- Department of Genetics, University of Calcutta, University College of Science (UCSTA), 35, Ballygunge Circular Road, Kolkata, 700 019, India
- Cactus Communications, Mumbai, India
| | - Sayak Ganguli
- Department of Biotechnology, St. Xavier's College (Autonomous), 30, Mother Teresa Sarani, Kolkata, 700 016, India.
| | - Mainak Sengupta
- Department of Genetics, University of Calcutta, University College of Science (UCSTA), 35, Ballygunge Circular Road, Kolkata, 700 019, India.
| |
Collapse
|
16
|
Gan Q, Shao J, Cao Y, Lei J, Xie P, Ge J, Hu G. USP33 regulates c-Met expression by deubiquitinating SP1 to facilitate metastasis in hepatocellular carcinoma. Life Sci 2020; 261:118316. [PMID: 32835698 DOI: 10.1016/j.lfs.2020.118316] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/05/2020] [Accepted: 08/19/2020] [Indexed: 12/21/2022]
Abstract
AIMS Deubiquitinase ubiquitin-specific protease 33 (USP33) is abnormally expressed in various tumors and participates in tumor progression. However, the expression and biological role of USP33 in hepatocellular carcinoma (HCC) are still unclear. MAIN METHODS We performed immunohistochemistry, western blotting, and qRT-PCR analysis to determine the expression of USP33 in HCC. We then analyzed the effects of USP33 expression on the prognosis of HCC. The roles of USP33 in regulating HCC cell migration and invasion were further explored in vitro. Animal studies were performed to investigate the effects of USP33 on tumor metastasis. RNA sequencing and luciferase reporter and immunofluorescence assays were used to identify the activation of the specificity protein 1 (SP1)/c-Met axis. KEY FINDINGS Here, for the first time, we reported an abnormal increase in the expression of USP33 in HCC tissues and that USP33 may act as a prognostic biomarker for HCC patients. We found that USP33 knockdown inhibited the invasion and metastasis in HCC cells both in vitro and in vivo, which was partly dependent on c-Met. Further investigations revealed that USP33 regulated c-Met expression by enhancing the protein stability of the transcription factor SP1 in HCC cells. Mechanistically, USP33 directly bound SP1 and decreased its ubiquitination, thereby upregulating c-Met expression. SIGNIFICANCE Our results reveal that USP33 acts as the deubiquitinating enzyme of SP1 and contributes to HCC invasion and metastasis through activation of the SP1/c-Met axis. These data indicate a previously unknown function of USP33, which may provide potential targets for the treatment of HCC patients.
Collapse
Affiliation(s)
- Qin Gan
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jiujiang Hospital of Nanchang University, Jiujiang 332000, China; Department of Hepatobiliary and Pancreatic Surgery, Jiujiang NO.1 People's Hospital, Jiujiang 332000, China
| | - Jia Shao
- Centre for Assisted Reproduction, The First Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Yan Cao
- Department of Gastroenterology, The Fourth Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jun Lei
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Peiyi Xie
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jin Ge
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China.
| | - Guohui Hu
- Department of General Practice, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China.
| |
Collapse
|
17
|
Wolf J, Auw-Haedrich C, Schlecht A, Boneva S, Mittelviefhaus H, Lapp T, Agostini H, Reinhard T, Schlunck G, Lange CAK. Transcriptional characterization of conjunctival melanoma identifies the cellular tumor microenvironment and prognostic gene signatures. Sci Rep 2020; 10:17022. [PMID: 33046735 PMCID: PMC7550331 DOI: 10.1038/s41598-020-72864-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023] Open
Abstract
This study characterizes the transcriptome and the cellular tumor microenvironment (TME) of conjunctival melanoma (CM) and identifies prognostically relevant biomarkers. 12 formalin-fixed and paraffin-embedded CM were analyzed by MACE RNA sequencing, including six cases each with good or poor clinical outcome, the latter being defined by local recurrence and/or systemic metastases. Eight healthy conjunctival specimens served as controls. The TME of CM, as determined by bioinformatic cell type enrichment analysis, was characterized by the enrichment of melanocytes, pericytes and especially various immune cell types, such as plasmacytoid dendritic cells, natural killer T cells, B cells and mast cells. Differentially expressed genes between CM and control were mainly involved in inhibition of apoptosis, proteolysis and response to growth factors. POU3F3, BIRC5 and 7 were among the top expressed genes associated with inhibition of apoptosis. 20 genes, among them CENPK, INHA, USP33, CASP3, SNORA73B, AAR2, SNRNP48 and GPN1, were identified as prognostically relevant factors reaching high classification accuracy (area under the curve: 1.0). The present study provides new insights into the TME and the transcriptional profile of CM and additionally identifies new prognostic biomarkers. These results add new diagnostic tools and may lead to new options of targeted therapy for CM.
Collapse
Affiliation(s)
- Julian Wolf
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Claudia Auw-Haedrich
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Anja Schlecht
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Stefaniya Boneva
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Hans Mittelviefhaus
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Thabo Lapp
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Hansjürgen Agostini
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Thomas Reinhard
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Günther Schlunck
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany
| | - Clemens A K Lange
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106, Freiburg, Germany.
| |
Collapse
|
18
|
Deubiquitinating enzyme USP33 restrains docetaxel-induced apoptosis via stabilising the phosphatase DUSP1 in prostate cancer. Cell Death Differ 2019; 27:1938-1951. [PMID: 31857702 DOI: 10.1038/s41418-019-0473-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 11/09/2022] Open
Abstract
The treatment of castration-resistant prostate cancer (CRPC) still faces many challenges. Docetaxel is a chemotherapeutic drug commonly used in CRPC patients. However, docetaxel-based chemotherapy usually causes docetaxel resistance, partially due to the resistance of CRPC cells to docetaxel-induced apoptosis. Here, we report that the deubiquitinating enzyme ubiquitin-specific protease 33 (USP33) inhibits docetaxel-induced apoptosis of prostate cancer cells, including androgen-independent prostate cancer cells. USP33 is overexpressed in prostate cancer cells and tissues. We found that knockdown or knockout of USP33 enhanced docetaxel-induced apoptosis of prostate cancer cells, accompanied by increased phosphorylation of the cJUN NH2-terminal kinase (JNK). After blocking docetaxel-induced JNK activation using the JNK inhibitor SP600125 or siRNA targeting JNK, the USP33 knockout-enhanced apoptosis was reversed. Furthermore, we found that USP33 could interact with the phosphatase DUSP1 to negatively regulate the activation of JNK, while USP33 knockdown promoted the proteasomal degradation of DUSP1. Mechanistically, we found that USP33 could inhibit the Lys48 (K48)-linked polyubiquitination of DUSP1. More importantly, DUSP1 overexpression could reverse the USP33 knockdown-induced JNK activation and apoptosis in docetaxel-treated prostate cancer cells. Therefore, USP33 overexpression in prostate cancer may contribute to docetaxel resistance by inhibiting the degradation of its partner DUSP1, leading to impaired JNK activation and apoptosis. Our study suggests that USP33-DUSP1-JNK may be a key signalling module mediating the docetaxel resistance of CRPC, indicating that USP33 is a potential novel therapeutic target in CRPC.
Collapse
|
19
|
Functional analysis of deubiquitylating enzymes in tumorigenesis and development. Biochim Biophys Acta Rev Cancer 2019; 1872:188312. [DOI: 10.1016/j.bbcan.2019.188312] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/16/2019] [Accepted: 08/16/2019] [Indexed: 02/06/2023]
|
20
|
Niu K, Fang H, Chen Z, Zhu Y, Tan Q, Wei D, Li Y, Balajee AS, Zhao Y. USP33 deubiquitinates PRKN/parkin and antagonizes its role in mitophagy. Autophagy 2019; 16:724-734. [PMID: 31432739 DOI: 10.1080/15548627.2019.1656957] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
PRKN/parkin activation through phosphorylation of its ubiquitin and ubiquitin-like domain by PINK1 is critical in mitophagy induction for eliminating the damaged mitochondria. Deubiquitinating enzymes (DUBs) functionally reversing PRKN ubiquitination are critical in controlling the magnitude of PRKN-mediated mitophagy process. However, potential DUBs that directly target PRKN and antagonize its pro-mitophagy effect remains to be identified and characterized. Here, we demonstrated that USP33/VDU1 is localized at the outer membrane of mitochondria and serves as a PRKN DUB through their interaction. Cellular and in vitro assays illustrated that USP33 deubiquitinates PRKN in a DUB activity-dependent manner. USP33 prefers to remove K6, K11, K48 and K63-linked ubiquitin conjugates from PRKN, and deubiquitinates PRKN mainly at Lys435. Mutation of this site leads to a significantly decreased level of K63-, but not K48-linked PRKN ubiquitination. USP33 deficiency enhanced both K48- and K63-linked PRKN ubiquitination, but only K63-linked PRKN ubiquitination was significantly increased under mitochondrial depolarization. Further, USP33 knockdown increased both PRKN protein stabilization and its translocation to depolarized mitochondria leading to the enhancement of mitophagy. Moreover, USP33 silencing protects SH-SY5Y human neuroblastoma cells from the neurotoxin MPTP-induced apoptotic cell death. Our findings convincingly demonstrate that USP33 is a novel PRKN deubiquitinase antagonizing its regulatory roles in mitophagy and SH-SY5Y neuron-like cell survival. Thus, USP33 inhibition may represents an attractive new therapeutic strategy for PD patients.Abbreviations: CCCP: carbonyl cyanide 3-chlorophenylhydrazone; DUB: deubiquitinating enzymes; MPTP: 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; OMM: outer mitochondrial membrane; PD: Parkinson disease; PINK1: PTEN induced kinase 1; PRKN/PARK2: parkin RBR E3 ubiquitin protein ligase; ROS: reactive oxygen species; TM: transmembrane; Ub: ubiquitin; UBA1: ubiquitin like modifier activating enzyme 1; UBE2L3/UbcH7: ubiquitin conjugating enzyme E2 L3; USP33: ubiquitin specific peptidase 33; WT: wild type.
Collapse
Affiliation(s)
- Kaifeng Niu
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hongbo Fang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Zixiang Chen
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuqi Zhu
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qunsong Tan
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Di Wei
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Yueyang Li
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Adayabalam S Balajee
- REAC/TS, Oak Ridge Associated Universities, Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | - Yongliang Zhao
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
21
|
Jiang Z, Liang G, Xiao Y, Qin T, Chen X, Wu E, Ma Q, Wang Z. Targeting the SLIT/ROBO pathway in tumor progression: molecular mechanisms and therapeutic perspectives. Ther Adv Med Oncol 2019; 11:1758835919855238. [PMID: 31217826 PMCID: PMC6557020 DOI: 10.1177/1758835919855238] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/07/2019] [Indexed: 01/14/2023] Open
Abstract
The SLITs (SLIT1, SLIT2, and SLIT3) are a family of secreted proteins that mediate positional interactions between cells and their environment during development by signaling through ROBO receptors (ROBO1, ROBO2, ROBO3, and ROBO4). The SLIT/ROBO signaling pathway has been shown to participate in axonal repulsion, axon guidance, and neuronal migration in the nervous system and the formation of the vascular system. However, the role of the SLIT/ROBO pathway has not been thoroughly clarified in tumor development. The SLIT/ROBO pathway can produce both beneficial and detrimental effects in the growth of malignant cells. It has been confirmed that SLIT/ROBO play contradictory roles in tumorigenesis. Here, we discuss the tumor promotion and tumor suppression roles of the SLIT/ROBO pathway in tumor growth, angiogenesis, migration, and the tumor microenvironment. Understanding these roles will help us develop more effective cancer therapies.
Collapse
Affiliation(s)
- Zhengdong Jiang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Gang Liang
- Department of Hepatobiliary Surgery, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, Shaanxi, China
| | - Ying Xiao
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tao Qin
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xin Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Erxi Wu
- Department of Neurosurgery, Neuroscience Institute, Baylor Scott and White Health, Temple, TX, USA
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
22
|
Tong M, Jun T, Nie Y, Hao J, Fan D. The Role of the Slit/Robo Signaling Pathway. J Cancer 2019; 10:2694-2705. [PMID: 31258778 PMCID: PMC6584916 DOI: 10.7150/jca.31877] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/28/2019] [Indexed: 12/25/2022] Open
Abstract
The Slit family is a family of secreted proteins that play important roles in various physiologic and pathologic activities via interacting with Robo receptors. Slit/Robo signaling was first identified in the nervous system, where it functions in neuronal axon guidance; nevertheless, an increasing number of studies have shown that Slit/Robo signaling even regulates other activities, such as angiogenesis, inflammatory cell chemotaxis, tumor cell migration and metastasis. Although the precise role of the ligand-receptor in organisms has been obscure and the conclusions drawn are sometimes paradoxical, tremendous advances in understanding the Slit/Robo signaling pathway have been made. As such, our review summarizes the characteristics of the Slit/Robo signaling pathway and its role in various cell types.
Collapse
Affiliation(s)
- Mingfu Tong
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Tie Jun
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jianyu Hao
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
23
|
Xia Y, Wang L, Xu Z, Kong R, Wang F, Yin K, Xu J, Li B, He Z, Wang L, Xu H, Zhang D, Yang L, Wu JY, Xu Z. Reduced USP33 expression in gastric cancer decreases inhibitory effects of Slit2-Robo1 signalling on cell migration and EMT. Cell Prolif 2019; 52:e12606. [PMID: 30896071 PMCID: PMC6536419 DOI: 10.1111/cpr.12606] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/26/2019] [Accepted: 02/26/2019] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES Gastric cancer (GC) is one of the most common cancers in the world, causing a large number of deaths every year. The Slit-Robo signalling pathway, initially discovered for its critical role in neuronal guidance, has recently been shown to modulate tumour invasion and metastasis in several human cancers. However, the role of Slit-Robo signalling and the molecular mechanisms underlying its role in the pathogenesis of gastric cancer remains to be elucidated. MATERIALS AND METHODS Slit2, Robo1 and USP33 expressions were analysed in datasets obtained from the Oncomine database and measured in human gastric cancer specimens. The function of Slit2-Robo1-USP33 signalling on gastric cancer cells migration and epithelial-mesenchymal transition (EMT) was studied both in vitro and in vivo. The mechanism of the interaction between Robo1 and USP33 was explored by co-IP and ubiquitination protein analysis. RESULTS The mRNA and protein levels of Slit2 and Robo1 are lower in GC tissues relative to those in adjacent healthy tissues. Importantly, Slit2 inhibits GC cell migration and suppresses EMT process in a Robo-dependent manner. The inhibitory function of Slit2-Robo1 is mediated by ubiquitin-specific protease 33 (USP33) via deubiquitinating and stabilizing Robo1. USP33 expression is decreased in GC tissues, and reduced USP33 level is correlated with poor patient survival. CONCLUSIONS Our study reveals the inhibitory function of Slit-Robo signalling in GC and uncovers a role of USP33 in suppressing cancer cell migration and EMT by enhancing Slit2-Robo1 signalling. USP33 represents a feasible choice as a prognostic biomarker for GC.
Collapse
MESH Headings
- Aged
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Line, Tumor
- Cell Movement
- Down-Regulation
- Epithelial-Mesenchymal Transition
- Female
- Gene Expression Regulation, Neoplastic
- Heterografts
- Humans
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Middle Aged
- Models, Biological
- Neoplasm Transplantation
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Prognosis
- Protein Stability
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/genetics
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Signal Transduction
- Stomach Neoplasms/genetics
- Stomach Neoplasms/metabolism
- Stomach Neoplasms/pathology
- Ubiquitin Thiolesterase/antagonists & inhibitors
- Ubiquitin Thiolesterase/genetics
- Ubiquitin Thiolesterase/metabolism
- Ubiquitination
- Roundabout Proteins
Collapse
Affiliation(s)
- Yiwen Xia
- Department of Gastric SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Linjun Wang
- Department of Gastric SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Zhipeng Xu
- Department of Gastric SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Ruirui Kong
- State Key Laboratory of Brain and Cognitive Science, Institute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Fei Wang
- State Key Laboratory of Brain and Cognitive Science, Institute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Kai Yin
- Department of General SurgeryAffiliated Hospital of Jiangsu UniversityZhenjiangChina
| | - Jianghao Xu
- Department of Gastric SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Bowen Li
- Department of Gastric SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Zhongyuan He
- Department of Gastric SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Lu Wang
- Department of Gastric SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Hao Xu
- Department of Gastric SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Diancai Zhang
- Department of Gastric SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Li Yang
- Department of Gastric SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Jane Y. Wu
- State Key Laboratory of Brain and Cognitive Science, Institute of BiophysicsChinese Academy of SciencesBeijingChina
- Department of Neurology, Center for Genetic MedicineNorthwestern University Feinberg School of MedicineChicagoIllinois
- Department of NeurologyCenter for Genetic MedicineLurie Cancer CenterChicagoIllinois
| | - Zekuan Xu
- Department of Gastric SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and TreatmentJiangsu Collaborative Innovation Center for Cancer Personalized MedicineSchool of Publich HealthNanjing Medical UniversityNanjingChina
| |
Collapse
|
24
|
Xu ZH, Wang H, Ji XY, Zhang FX, Gao BL, Hu JA, Zheng J. A detrimental mutation on USP40 unlocks the tumorigenesis in a rare case of lung cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:740-749. [PMID: 31933881 PMCID: PMC6945189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 11/20/2018] [Indexed: 06/10/2023]
Abstract
Lung adenocarcinoma (LUAD) is the most common histologic subtype of lung cancer. Previous research has shown heterogeneity in lung cancer, with the parallel existence of multiple subclones characterized by their own specific mutational landscape. The aim of our study was to gain insight into the evolutionary pattern of lung cancer by investigating the genomic heterogeneity between a nodule and its distant tumor. Luckily, we obtained nodule and tumor samples derived from surgery and a blood sample from a single patient. The samples are very unique, for tissues with the same genetic background from nodules to malignant tumors are rarely available and require precise micro-cutting. In this study, we performed whole-genome sequencing of these two samples, to identify novel candidate driver genes associated with LUAD. The nodule and tumor were found to have common significant ubiquitin-specific protease 40 (USP40) mutations, indicating an important driver role for the gene. Moreover, we also observed the two novel candidate driver genes ASCL5 and CAPNS1 in the LUAD sample. In summary, we pinpoint the predominant mutations in LUAD by WES, highlighting the substantial genetic alterations contributing to LUAD tumorigenesis. This may provide a better understanding of the clonal evolution during tumor development.
Collapse
Affiliation(s)
- Zhi-Hong Xu
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiaotong UniversityShanghai, P. R. China
| | - Hui Wang
- Laboratory of Molecular Neuropharmacology, School of Pharmacy East China University of Science and TechnologyShanghai, P. R. China
| | - Xiao-Yang Ji
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiaotong UniversityShanghai, P. R. China
| | - Fei-Xu Zhang
- Laboratory of Molecular Neuropharmacology, School of Pharmacy East China University of Science and TechnologyShanghai, P. R. China
| | - Bei-Li Gao
- Department of Respiratory Medicine, Ruijin Hospital, Shanghai Jiaotong UniversityShanghai, P. R. China
| | - Jia-An Hu
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiaotong UniversityShanghai, P. R. China
| | - Jing Zheng
- Laboratory of Molecular Neuropharmacology, School of Pharmacy East China University of Science and TechnologyShanghai, P. R. China
| |
Collapse
|
25
|
Shen X, Niu C, Guo J, Xia M, Xia J, Hu Y, Zheng Y. Stra8 may inhibit apoptosis during mouse spermatogenesis via the AKT signaling pathway. Int J Mol Med 2018; 42:2819-2830. [PMID: 30106128 DOI: 10.3892/ijmm.2018.3825] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 08/02/2018] [Indexed: 11/06/2022] Open
Abstract
Stimulated by retinoic acid 8 (Stra8), one of genes induced by retinoic acid (RA), is required for the meiotic initiation of male spermatogenesis. The present study found that Stra8 inhibited apoptosis in male Stra8‑knockout mice, and in mice with vitamin A deficiency and vitamin A recovery in vivo. This phenotype was also verified in GC1 spermatogonia (spg) cells overexpressing Stra8. In addition, microarray analysis identified that there were nine differentially expressed genes (DEGs) in the Stra8‑overexpressed GC1 spg cells compared with the control groups; the expression of these nine genes was verified via mRNA expression levels. The DEGs were as follows: Phosphatidylinositol‑dependent kinase 1 (PDK1), a key gene upstream of protein kinase B (AKT); angiopoietin 2, a B‑cell lymphoma 2 (Bcl‑2)‑inhibited gene; transcription factor 4, glutathione S‑transferase P91 and ubiquitin‑specific protease 33, mitogen‑activated protein kinase (MAPK)‑related genes; oxidative stress induced growth inhibitor 1, related to the P53 pathway; Bcl‑2, P53, ERK (MAPK1/3), c‑Jun N‑terminal kinase (MAPK8/9), and P38 (MAPK14), all of which are key genes involved in the AKT signaling pathway. Therefore, the present study further verified these genes and found that the mRNA and protein expression levels of PDK1, AKT, Bcl‑2 and ERK were increased. Although the mRNA expression level of P53 was decreased, there was no significant difference in the protein expression level in Stra8‑overexpressing GC1 spg cells compared with controls. In addition, Caspase 3, one of the executioner caspases, was decreased in Stra8‑overexpressing GC1 spg cells compared with the control groups. Therefore, it was suggested that Stra8 may directly or indirectly inhibit caspases through the AKT signaling pathway and ultimately exert an anti‑apoptotic effect in the male reproductive system.
Collapse
Affiliation(s)
- Xueyi Shen
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Changmin Niu
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Jiaqian Guo
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Mengmeng Xia
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Jing Xia
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Yanqiu Hu
- Center of Reproductive Medicine, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Ying Zheng
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| |
Collapse
|
26
|
Mackin SJ, O'Neill KM, Walsh CP. Comparison of DNMT1 inhibitors by methylome profiling identifies unique signature of 5-aza-2'deoxycytidine. Epigenomics 2018; 10:1085-1101. [PMID: 30070602 DOI: 10.2217/epi-2017-0171] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
AIM 5-aza-2'deoxycytidine (Aza) is used to treat myelodysplastic syndrome and is in trials for other cancers. It acts chiefly as a hypomethylating agent inhibiting DNMT1. A lack of understanding of off-target effects in normal cells hinders wider usage. MATERIALS & METHODS We compared treatment of the same normosomic, nontransformed fibroblast cell line with Aza and SMARTpool siRNA against DNMT1. Methylation and transcription were assayed using Illumina 450k and HT12 arrays. RESULTS Both Aza and DNMT1 siRNA caused overall hypomethylation, with siRNA more efficient at demethylating gene bodies. Hypomethylation at the promoters of many histones, and hypermethylation at multiple sites genome wide, were unique to Aza treatment. CONCLUSION Aza had important unique effects and targets compared with DNMT1 inhibition via siRNA.
Collapse
Affiliation(s)
- Sarah-Jayne Mackin
- Genomic Medicine Research Group, Centre for Molecular Biosciences, Biomedical Sciences Research Institute, Ulster University, Coleraine, BT52 1SA, UK
| | - Karla M O'Neill
- Genomic Medicine Research Group, Centre for Molecular Biosciences, Biomedical Sciences Research Institute, Ulster University, Coleraine, BT52 1SA, UK.,Current address: The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7AE, UK
| | - Colum P Walsh
- Genomic Medicine Research Group, Centre for Molecular Biosciences, Biomedical Sciences Research Institute, Ulster University, Coleraine, BT52 1SA, UK
| |
Collapse
|
27
|
Yan M, Zhao C, Wei N, Wu X, Cui J, Xing Y. High Expression of Ubiquitin-Specific Protease 8 (USP8) Is Associated with Poor Prognosis in Patients with Cervical Squamous Cell Carcinoma. Med Sci Monit 2018; 24:4934-4943. [PMID: 30010158 PMCID: PMC6067021 DOI: 10.12659/msm.909235] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/27/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Cervical cancer is one of the most common female malignancies in the world. The ubiquitin-specific protease 8 (USP8) functions by removing ubiquitin from protein substrates, and its potential role in cancer development was recently uncovered in lung cancer. The aim of this study was to investigate the expression and function of USP8 in cervical squamous cell carcinoma (CSCC). MATERIAL AND METHODS Immunohistochemical staining and quantitative PCR were performed to explore the expression of USP8 in both CSCC tissues and adjacent normal cervical tissues. Univariate and multivariate analyses were conducted to evaluate the clinical significance of USP8 in CSCC. Proliferation, migration, and invasion abilities of 2 CSCC cell lines were assessed after overexpression or silencing USP8, respectively. RESULTS Both the RNA and protein levels of USP8 were upregulated in CSCC tissues compared to normal cervical tissues. High expression of USP8 was correlated with advanced tumor stage and high recurrence risk. Moreover, USP8 was identified as a novel independent prognostic factor for CSCC patients. Cellular studies showed that USP8 can enhance the proliferation, migration, and invasion abilities of CSCC cells, thereby promoting tumor progression. CONCLUSIONS High expression of USP8 is frequent in CSCC tissues, which promotes tumor proliferation and invasion, and is correlated with a poor overall survival. Targeting USP8 may be a novel direction for drug development for CSCC therapy.
Collapse
Affiliation(s)
- Min Yan
- Department of Obstetrics, Yidu Central Hospital of Weifang, Weifang, Shandong, P.R. China
| | - Cuihong Zhao
- Department of Obstetrics, Yidu Central Hospital of Weifang, Weifang, Shandong, P.R. China
| | - Na Wei
- Department of Obstetrics, Yidu Central Hospital of Weifang, Weifang, Shandong, P.R. China
| | - Xiaoqian Wu
- Department of Cardiology, Yidu Central Hospital of Weifang, Weifang, Shandong, P.R. China
| | - Jianli Cui
- Family Planning Station of Qingzhou, Weifang, Shandong, P.R. China
| | - Yanling Xing
- Department of Obstetrics and Gynecology, Nangang Branch of Heilongjiang Province Hospital, Harbin, Heilongjiang, P.R. China
| |
Collapse
|
28
|
Chen Y, Pang X, Ji L, Sun Y, Ji Y. Reduced Expression of Deubiquitinase USP33 Is Associated with Tumor Progression and Poor Prognosis of Gastric Adenocarcinoma. Med Sci Monit 2018; 24:3496-3505. [PMID: 29802710 PMCID: PMC5996837 DOI: 10.12659/msm.908075] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background Ubiquitin-specific peptidase 33 (USP33) is a deubiquitinase that balances the ubiquitin status of proteins. It has been reported to act as a tumor suppressor in colorectal cancer and lung cancer. However, the expression pattern and clinical significance of USP33 have not been investigated in gastric adenocarcinoma (GAC). Material/Methods We explored the USP33 protein and RNA levels by immunohistochemistry (IHC), Western blot analysis, and qRT-PCR. The Pearson chi-square test was performed to evaluate the statistical associations between USP33 level and patient characteristics. Additionally, the relationship between USP33 expression and patient survival was investigated. Cellular studies, including proliferation assay, migration assay, and invasion assay, were conducted to demonstrate the underlying mechanisms of USP33 in GAC progression. Results This study included 121 patients with GAC. USP33 showed a decreased expression in GAC tissues compared to adjacent normal gastric tissues. Low expression of USP33 was correlated with invasion depth and advanced TNM stage. According to survival analysis, upper location of tumor (P=0.003), invasion depth (P=0.048), advanced TNM stage (P=0.001), and low USP33 level (P=0.001) were all associated with poor overall survival of GAC patients. Cox analysis confirmed the independent role of USP33 in predicting patient survival. Cell experiments showed that USP33 overexpression significantly inhibited the proliferation, migration, and invasion of GAC cells. Conclusions USP33 was downregulated in GAC, and was an independent prognostic factor. In vitro results demonstrated the role of USP33 in suppressing tumor progression, suggesting that the developing an agonist of USP33 may be a novel direction for chemotherapy development.
Collapse
Affiliation(s)
- Yan Chen
- Department of Gastroenterology, Yidu Central Hospital of Weifang, Weifang, Shandong, China (mainland)
| | - Xumei Pang
- Department of Oncology, Yidu Central Hospital of Weifang, Weifang, Shandong, China (mainland)
| | - Lijuan Ji
- Department of Gastroenterology, Yidu Central Hospital of Weifang, Weifang, Shandong, China (mainland)
| | - Yingchun Sun
- Department of Neurology, Shouguang Hospital of Traditional Chinese Medicine, Weifang, Shandong, China (mainland)
| | - Yongjing Ji
- Jinan Second People's Hospital (The Ophthalmologic Hospital of Jinan), Jinan, Shandong, China (mainland)
| |
Collapse
|
29
|
Wang Y, Zhang S, Bao H, Mu S, Zhang B, Ma H, Ma S. MicroRNA-365 promotes lung carcinogenesis by downregulating the USP33/SLIT2/ROBO1 signalling pathway. Cancer Cell Int 2018; 18:64. [PMID: 29743814 PMCID: PMC5930950 DOI: 10.1186/s12935-018-0563-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 04/21/2018] [Indexed: 12/19/2022] Open
Abstract
Background Abnormal microRNA expression is closely related to cancer occurrence and development. miR-365a-3p plays an oncogenic role in skin cancer, but its role in lung cancer remains unclear. In this study, we aimed to investigate its role and underlying molecular mechanisms in lung cancer. Methods Western blot and real-time quantitative PCR (qPCR) were used to detect the expression of miR-365a-3p in lung adenocarcinoma and lung cancer cell lines. The effects of miR-365a-3p on lung cancer cell proliferation, migration, and invasion were also explored in vitro. The potential miR-365a-3p that targets USP33 was determined by dual luciferase reporter assay and verified by qPCR and western blot analysis. miR-365a-3p acts as an oncogene by promoting lung carcinogenesis via the downregulation of the miR-365a/USP33/SLIT2/ROBO1 axis based on western blot analysis. Subcutaneous tumourigenesis further demonstrated that miR-365a-3p promotes tumour formation in vivo. Results miR-365a-3p was upregulated in lung adenocarcinoma and lung cancer cell lines. Overexpression of miR-365a-3p promoted and inhibition of miR-365a-3p suppressed the proliferation, migration, and invasion of lung cancer cells. We identified USP33 as the downstream target of miR-365a-3p and observed a negative correlation between miR-365a-3p and USP33 expression in lung adenocarcinoma patients. The miR-365/USP33/SLIT2/ROBO1 axis, a new mechanism, was reported to inhibit the invasion and metastasis of lung cancer. A nude mouse model of lung cancer further verified these findings. Conclusions In summary, miR-365a-3p acts as an oncogene by promoting lung carcinogenesis via the downregulation of the USP33/SLIT2/ROBO1 signalling pathway, making the miR-365/USP33/SLIT2/ROBO1 axis a new mechanism of lung cancer promotion and a novel therapeutic target for predicting prognosis and response to gene therapy. Electronic supplementary material The online version of this article (10.1186/s12935-018-0563-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuhuan Wang
- 1Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 Guangdong China
| | - Shuhua Zhang
- 1Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 Guangdong China
| | - Hejing Bao
- Department of Oncology, Chongqing Three Gorges Center Hospital, Chongqing, China
| | - Shukun Mu
- 1Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 Guangdong China
| | - Baishen Zhang
- 1Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 Guangdong China
| | - Hao Ma
- 3Department of Clinical Medicine, Tianjin Medical University College, Tianjin, China
| | - Shudong Ma
- 1Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 Guangdong China
| |
Collapse
|
30
|
Liu H, Zhang Q, Li K, Gong Z, Liu Z, Xu Y, Swaney MH, Xiao K, Chen Y. Prognostic significance of USP33 in advanced colorectal cancer patients: new insights into β-arrestin-dependent ERK signaling. Oncotarget 2018; 7:81223-81240. [PMID: 27835898 PMCID: PMC5348388 DOI: 10.18632/oncotarget.13219] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/14/2016] [Indexed: 01/05/2023] Open
Abstract
Patients with liver metastases of colorectal cancer (CRCLM) have a poorer prognosis compared to colorectal cancer (CRC) patients in local stage. Evaluating the recurrence and overall survival of advanced patients is critical in improving disease treatment and clinical outcome. Here we investigated the expression pattern of USP33, a deubiquitinating enzyme, in both primary CRC tissues and liver metastases tissues. Univariate and multivariate analyses identified that low expression of USP33 in CRCLM tissues indicated high recurrence risk and poor overall prognosis. Overexpression of USP33 can significantly inhibit cell proliferation, migration, and invasion. On the other hand, USP33 knock-down promoted cell proliferation and invasion under SDF-1 stimulation; whereas dynasore (an internalization inhibitor) pretreatment in USP33 silencing cells showed a distinct antipromoting effect, revealing the participation of CXCR4 internalization in regulating tumor progress. Further results verified that USP33 can deubiquitinate β-arrestin2, subsequently block the internalization of SDF-1-stimulated CXCR4, and disrupt β-arrestin-dependent ERK activation. The existence and functions of β-arrestin-dependent signaling have been previously determined in several Gs-coupled receptors, such as β2-adrenergic receptor and angiotensin receptor subtype 1a; however, little is known about this in Gi-coupled receptors. Our study not only established USP33 as a novel prognosis biomarker in advanced CRCLM patients, but also highlighted the significance of β-arrestin-dependent ERK signaling in cancer development.
Collapse
Affiliation(s)
- Hongda Liu
- Department of General Surgery, Qilu Hospital Affiliated to Shandong University, Jinan, Shandong 250012, China.,Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, Shandong 250012, China.,Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Qun Zhang
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China
| | - Kangshuai Li
- Department of General Surgery, Qilu Hospital Affiliated to Shandong University, Jinan, Shandong 250012, China
| | - Zheng Gong
- Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zhaochen Liu
- Department of General Surgery, Qilu Hospital Affiliated to Shandong University, Jinan, Shandong 250012, China
| | - Yunfei Xu
- Department of General Surgery, Qilu Hospital Affiliated to Shandong University, Jinan, Shandong 250012, China
| | - Mary Hannah Swaney
- Pennsylvania State University, University Park, Pennsylvania, 16802, USA
| | - Kunhong Xiao
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Yuxin Chen
- Department of General Surgery, Qilu Hospital Affiliated to Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
31
|
Cheng Q, Yuan Y, Li L, Guo T, Miao Y, Ren Y, Liu J, Feng Q, Wang X, Zhao P, Zuo Y, Qian L, Zhang L, Zheng H. Deubiquitinase USP33 is negatively regulated by β-TrCP through ubiquitin-dependent proteolysis. Exp Cell Res 2017; 356:1-7. [PMID: 28506875 DOI: 10.1016/j.yexcr.2017.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 05/03/2017] [Accepted: 05/12/2017] [Indexed: 10/19/2022]
Abstract
Ubiquitin-mediated proteolysis regulates cellular levels of various proteins, and therefore plays important roles in controlling cell signaling and disease progression. The Skp1-Cul1-F-box ubiquitin ligase β-TrCP is recognized as an important negative regulator for numerous key signaling proteins. Recently, the deubiquitinases (DUBs) have turned out to be essential to regulate signaling pathways related to human diseases. However, whether β-TrCP is able to regulate the deubiquitinase family members remains largely unexplored. Here, we found that β-TrCP downregulated cellular levels of endogenous USP33. We also revealed that β-TrCP interacted with USP33 independently of the classic binding motif for β-TrCP, and mediated USP33 degradation via the ubiquitin proteasome pathway. Furthermore, we found that the WD40 motif of β-TrCP and 201-400 amino acid motif of USP33 are required for the interaction between β-TrCP and USP33. Consequently, β-TrCP attenuated USP33-mediated inhibition of cell proliferation and cell invasion. Taken together, our study clarified that the E3 ligase β-TrCP regulates cellular USP33 levels by the ubiquitin-proteasomal proteolysis.
Collapse
Affiliation(s)
- Qiao Cheng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Yukang Yuan
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Lemin Li
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Tingting Guo
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Ying Miao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Ying Ren
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Jin Liu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Qian Feng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Xiaofang Wang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Peng Zhao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Yibo Zuo
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Liping Qian
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Liting Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Hui Zheng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province 215123, China.
| |
Collapse
|
32
|
Ma M, Yu N. Over-Expression of TBL1XR1 Indicates Poor Prognosis of Serous Epithelial Ovarian Cancer. TOHOKU J EXP MED 2017; 241:239-247. [DOI: 10.1620/tjem.241.239] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Ming Ma
- Department of Oncology, Linyi People’s Hospital
| | - Nina Yu
- Department of Gynecology and Obstetrics, Linyi People’s Hospital
| |
Collapse
|
33
|
Huang T, Kang W, Cheng ASL, Yu J, To KF. The emerging role of Slit-Robo pathway in gastric and other gastro intestinal cancers. BMC Cancer 2015; 15:950. [PMID: 26674478 PMCID: PMC4682238 DOI: 10.1186/s12885-015-1984-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 12/08/2015] [Indexed: 01/12/2023] Open
Abstract
Gastric cancer remains one of the most common cancers worldwide and one of the leading cause for cancer-related deaths. Due to the high frequency of metastasis, it is still one of the most lethal malignancies in which kinds of signaling pathways are involved in. The Roundabout (ROBO) receptors and their secreted SLIT glycoprotein ligands, which were originally identified as important axon guidance molecules, have implication in the regulation of neurons and glia, leukocytes, and endothelial cells migration. Recent researches also put high emphasis on the important roles of the Slit-Robo pathway in tumorigenesis, cancer progression and metastasis. Herein we provide a comprehensive review on the role of these molecules and their associated signaling pathway in gastric and other gastrointestinal cancers. Improved knowledge of the Slit-Robo signaling pathway in gastric carcinoma will be useful for deep understanding the mechanisms of tumor development and identifying ideal targets of anticancer therapy in gastric carcinoma.
Collapse
Affiliation(s)
- Tingting Huang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, PR China.
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, PR China.
| | - Alfred S L Cheng
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, PR China.
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, PR China.
| | - Jun Yu
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, PR China.
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, PR China.
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, PR China.
| |
Collapse
|
34
|
Magraoui FE, Reidick C, Meyer HE, Platta HW. Autophagy-Related Deubiquitinating Enzymes Involved in Health and Disease. Cells 2015; 4:596-621. [PMID: 26445063 PMCID: PMC4695848 DOI: 10.3390/cells4040596] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/15/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
Autophagy is an evolutionarily-conserved process that delivers diverse cytoplasmic components to the lysosomal compartment for either recycling or degradation. This involves the removal of protein aggregates, the turnover of organelles, as well as the elimination of intracellular pathogens. In this situation, when only specific cargoes should be targeted to the lysosome, the potential targets can be selectively marked by the attachment of ubiquitin in order to be recognized by autophagy-receptors. Ubiquitination plays a central role in this process, because it regulates early signaling events during the induction of autophagy and is also used as a degradation-tag on the potential autophagic cargo protein. Here, we review how the ubiquitin-dependent steps of autophagy are balanced or counteracted by deubiquitination events. Moreover, we highlight the functional role of the corresponding deubiquitinating enzymes and discuss how they might be involved in the occurrence of cancer, neurodegenerative diseases or infection with pathogenic bacteria.
Collapse
Affiliation(s)
- Fouzi El Magraoui
- Biomedizinische Forschung, Human Brain Proteomics II, Leibniz-Institut für Analytische Wissenschaften - ISAS -e.V. 44139 Dortmund, Germany.
| | - Christina Reidick
- Biochemie Intrazellulärer Transportprozesse, Ruhr-Universität Bochum, 44801 Bochum, Germany.
| | - Hemut E Meyer
- Biomedizinische Forschung, Human Brain Proteomics II, Leibniz-Institut für Analytische Wissenschaften - ISAS -e.V. 44139 Dortmund, Germany.
| | - Harald W Platta
- Biochemie Intrazellulärer Transportprozesse, Ruhr-Universität Bochum, 44801 Bochum, Germany.
| |
Collapse
|
35
|
Liu C, Wang L, Chen W, Zhao S, Yin C, Lin Y, Jiang A, Zhang P. USP35 activated by miR let-7a inhibits cell proliferation and NF-κB activation through stabilization of ABIN-2. Oncotarget 2015; 6:27891-906. [PMID: 26348204 PMCID: PMC4695033 DOI: 10.18632/oncotarget.4451] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/16/2015] [Indexed: 12/22/2022] Open
Abstract
Ubiquitin specific protease 35 (USP35) is a member of deubiquitylases (DUBs). It remains largely unknown about the biological role and the regulation mechanism of USP35. Here, we first identified miR let-7a as a positive regulator of USP35 expression and showed that USP35 expression positively correlates with miR let-7a expression in different cancer cell lines and tissues. Then, we showed that USP35 expression was decreased dramatically in the tumor tissues compared with the adjacent non-cancerous tissues. USP35 overexpression inhibited cell proliferation in vitro and inhibited xenograft tumor growth in vivo. Furthermore, we revealed that USP35 acts as a functional DUB and stabilizes TNFAIP3 interacting protein 2 (ABIN-2) by promoting its deubiquitination. Functionally, both ABIN-2 and USP35 could inhibit TNFα-induced NF-κB activation and overexpression of ABIN-2 alleviated USP35-loss induced activation of NF-κB. Collectively, our data indicated that miR let-7a-regulated USP35 can inhibit NF-κB activation by deubiquitination and stabilization of ABIN-2 protein and eventually inhibit cell proliferation. Overall, our study provides a novel rationale of targeting miR let-7a-USP35-ABIN-2 pathway for the therapy of cancer patients.
Collapse
Affiliation(s)
- Chunyan Liu
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, 250012, P.R. China
| | - Lina Wang
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, 250012, P.R. China
| | - Weiwen Chen
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, 250012, P.R. China
| | - Shihu Zhao
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, 250012, P.R. China
| | - Chunli Yin
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, 250012, P.R. China
| | - Yani Lin
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, 250012, P.R. China
| | - Anli Jiang
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, 250012, P.R. China
| | - Pengju Zhang
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, 250012, P.R. China
| |
Collapse
|
36
|
Hu S, Lu Y, Orr B, Godek K, Mustachio LM, Kawakami M, Sekula D, Compton DA, Freemantle S, Dmitrovsky E. Specific CP110 Phosphorylation Sites Mediate Anaphase Catastrophe after CDK2 Inhibition: Evidence for Cooperation with USP33 Knockdown. Mol Cancer Ther 2015; 14:2576-85. [PMID: 26304236 DOI: 10.1158/1535-7163.mct-15-0443] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/19/2015] [Indexed: 11/16/2022]
Abstract
Chromosomal instability (CIN) is a hallmark of solid tumor biology and is implicated in carcinogenesis. Preferentially eliminating malignant cells by targeting CIN and aneuploidy is an attractive antineoplastic strategy. We previously reported that CDK2 antagonism causes lung cancer cells to undergo anaphase catastrophe and apoptosis through inhibition of phosphorylation of the centrosomal protein CP110. Cells with activating KRAS mutations were particularly sensitive to CDK2 inhibition due to downregulation of CP110 protein levels. This study investigated mechanisms of CDK2 antagonism that mediate anaphase catastrophe via changes in CP110 protein expression and how activated KRAS affects CP110 levels in lung cancers. Site-directed mutagenesis revealed candidate CDK phosphorylation sites of CP110 (residues Ser 170 and Thr 194) critical for conferring anaphase catastrophe by altering centrosome clustering in mitosis. Intriguingly, KRAS mutation can promote CP110 protein degradation by upregulating the ubiquitin ligase SCF(cyclinF), which targets CP110 protein for destabilization. Finally, CDK2 inhibitor response was enhanced when combined with knockdown of the deubiquitinase USP33 that in turn accelerates CP110 protein degradation. Thus, this study provides molecular pharmacologic insights into how CP110 expression regulates response to CDK2 inhibition. An improved understanding of in vitro antineoplastic mechanisms of combining CDK2 antagonism with induced CP110 repression provides a rationale for exploring clinical consequences of this strategy. Taken together, preclinical findings obtained from combining CDK2 inhibition with USP33 repression have implications for treating patients with non-small cell lung cancers.
Collapse
Affiliation(s)
- Shanhu Hu
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Yun Lu
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Bernardo Orr
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Kristina Godek
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Lisa Maria Mustachio
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Masanori Kawakami
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - David Sekula
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Duane A Compton
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Sarah Freemantle
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Ethan Dmitrovsky
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, and Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire.
| |
Collapse
|