1
|
Li H, Xu S, Li X, Wang P, Hu M, Li N, Zhou Q, Chang M, Yao S. Inhibition of the ITGB1 gene attenuates crystalline silica-induced pulmonary fibrosis via epithelial-mesenchymal transformation. Braz J Med Biol Res 2024; 57:e13486. [PMID: 39258668 PMCID: PMC11379350 DOI: 10.1590/1414-431x2024e13486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 07/24/2024] [Indexed: 09/12/2024] Open
Abstract
Silicosis is a systemic disease caused by long-term exposure to high concentrations of free silica dust particles in the workplace. It is characterized by a persistent inflammatory response, fibroblast proliferation, and excessive collagen deposition, leading to pulmonary interstitial fibrosis. Epithelial interstitial transformation (EMT) can cause epithelial cells to lose their tight junctions, cell polarity, and epithelial properties, thereby enhancing the properties of interstitial cells, which can lead to the progression of fibrosis and the formation of scar tissue. Integrin 1 (ITGB1) is considered an important factor for promoting EMT and tumor invasion in a variety of tumors and also plays an important role in the progression of fibrotic diseases. Therefore, ITGB1 can be used as a potential target for the treatment of silicosis. In this study, we found that silica exposure induced epithelial-mesenchymal transformation in rats and that the expression of integrin ITGB1 was elevated along with the EMT. We used CRISPR/Cas9 technology to construct integrin ITGB1 knockdown cell lines for in vitro experiments. We compared the expression of the EMT key proteins E-cadherin and vimentin in the ITGB1 knockdown cells and wild-type cells simultaneously stimulated by silica and detected the aggregation point distribution of E-cadherin and vimentin in the cells using laser confocal microscopy. Our results showed that ITGB1 knockout inhibited the ITGB1/ILK/Snail signaling pathway and attenuated the EMT occurrence compared to control cells. These results suggested that ITGB1 is associated with silica-induced EMT and may be a potential target for the treatment of silicosis.
Collapse
Affiliation(s)
- Haibin Li
- School of Public Health, North China University of Science and Technology, Tangshan, China
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Shushuo Xu
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Xinxiao Li
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Penghao Wang
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Meng Hu
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Ning Li
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Qiang Zhou
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Meiyu Chang
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Sanqiao Yao
- School of Public Health, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
2
|
Yan YH, Wei LL, Wu JW, Wei SQ, Jiang YY, Yu JL, Yang LL, Li GB. Discovering New Metallo-Deubiquitinase CSN5 Inhibitors by a Non-Catalytic Activity Assay Platform. J Med Chem 2024; 67:14649-14667. [PMID: 39129245 DOI: 10.1021/acs.jmedchem.4c01514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
COP9 signalosome catalytic subunit CSN5 plays a key role in tumorigenesis and tumor immunity, showing potential as an anticancer target. Currently, only a few CSN5 inhibitors have been reported, at least partially, due to the challenges in establishing assays for CSN5 deubiquitinase activity. Here, we present the establishment and validation of a simple and reliable non-catalytic activity assay platform for identifying CSN5 inhibitors utilizing a new fluorescent probe, CFP-1, that exhibits enhanced fluorescence and fluorescence polarization features upon binding to CSN5. By using this platform, we identified 2-aminothiazole-4-carboxylic acids as new CSN5 inhibitors, which inhibited CSN5 but slightly downregulated PD-L1 in cancer cells. Furthermore, through the integration of deep learning-enabled virtual screening, we discovered that shikonins are nanomolar CSN5 inhibitors, which can upregulate PD-L1 in HCT116 cells. The binding modes of these structurally distinct inhibitors with CSN5 were explored by using microsecond-scale molecular dynamics simulations and tryptophan quenching assays.
Collapse
Affiliation(s)
- Yu-Hang Yan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Liu-Liu Wei
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jing-Wei Wu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Si-Qi Wei
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ying-Ying Jiang
- College of Food and Bioengineering, Xihua University, Chengdu 610039, China
| | - Jun-Lin Yu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ling-Ling Yang
- College of Food and Bioengineering, Xihua University, Chengdu 610039, China
| | - Guo-Bo Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
3
|
Su C, Mo J, Dong S, Liao Z, Zhang B, Zhu P. Integrinβ-1 in disorders and cancers: molecular mechanisms and therapeutic targets. Cell Commun Signal 2024; 22:71. [PMID: 38279122 PMCID: PMC10811905 DOI: 10.1186/s12964-023-01338-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 09/27/2023] [Indexed: 01/28/2024] Open
Abstract
Integrinβ-1 (ITGB1) is a crucial member of the transmembrane glycoprotein signaling receptor family and is also central to the integrin family. It forms heterodimers with other ligands, participates in intracellular signaling and controls a variety of cellular processes, such as angiogenesis and the growth of neurons; because of its role in bidirectional signaling regulation both inside and outside the membrane, ITGB1 must interact with a multitude of substances, so a variety of interfering factors can affect ITGB1 and lead to changes in its function. Over the past 20 years, many studies have confirmed a clear causal relationship between ITGB1 dysregulation and cancer development and progression in a wide range of benign diseases and solid tumor types, which may imply that ITGB1 is a prognostic biomarker and a therapeutic target for cancer treatment that warrants further investigation. This review summarizes the biological roles of ITGB1 in benign diseases and cancers, and compiles the current status of ITGB1 function and therapy in various aspects of tumorigenesis and progression. Finally, future research directions and application prospects of ITGB1 are suggested. Video Abstract.
Collapse
Affiliation(s)
- Chen Su
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China
| | - Jie Mo
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China
| | - Shuilin Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China
| | - Zhibin Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, People's Republic of China.
| | - Peng Zhu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
4
|
Füzesi-Levi MG, Ben-Nissan G, Listov D, Fridmann Sirkis Y, Hayouka Z, Fleishman S, Sharon M. The C-terminal tail of CSNAP attenuates the CSN complex. Life Sci Alliance 2023; 6:e202201634. [PMID: 37460146 PMCID: PMC10355216 DOI: 10.26508/lsa.202201634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/20/2023] Open
Abstract
Protein degradation is one of the essential mechanisms that enables reshaping of the proteome landscape in response to various stimuli. The largest E3 ubiquitin ligase family that targets proteins to degradation by catalyzing ubiquitination is the cullin-RING ligases (CRLs). Many of the proteins that are regulated by CRLs are central to tumorigenesis and tumor progression, and dysregulation of the CRL family is frequently associated with cancer. The CRL family comprises ∼300 complexes, all of which are regulated by the COP9 signalosome complex (CSN). Therefore, CSN is considered an attractive target for therapeutic intervention. Research efforts for targeted CSN inhibition have been directed towards inhibition of the complex enzymatic subunit, CSN5. Here, we have taken a fresh approach focusing on CSNAP, the smallest CSN subunit. Our results show that the C-terminal region of CSNAP is tightly packed within the CSN complex, in a groove formed by CSN3 and CSN8. We show that a 16 amino acid C-terminal peptide, derived from this CSN-interacting region, can displace the endogenous CSNAP subunit from the complex. This, in turn, leads to a CSNAP null phenotype that attenuates CSN activity and consequently CRLs function. Overall, our findings emphasize the potential of a CSNAP-based peptide for CSN inhibition as a new therapeutic avenue.
Collapse
Affiliation(s)
- Maria G Füzesi-Levi
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Gili Ben-Nissan
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Dina Listov
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | - Zvi Hayouka
- Institute of Biochemistry, Food Science and Nutrition, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Sarel Fleishman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Sharon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
5
|
Xu M, Yang L, Lin Y, Lu Y, Bi X, Jiang T, Deng W, Zhang L, Yi W, Xie Y, Li M. Emerging nanobiotechnology for precise theranostics of hepatocellular carcinoma. J Nanobiotechnology 2022; 20:427. [PMID: 36175957 PMCID: PMC9524074 DOI: 10.1186/s12951-022-01615-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/31/2022] [Indexed: 11/18/2022] Open
Abstract
Primary liver cancer has become the second most fatal cancer in the world, and its five-year survival rate is only 10%. Most patients are in the middle and advanced stages at the time of diagnosis, losing the opportunity for radical treatment. Liver cancer is not sensitive to chemotherapy or radiotherapy. At present, conventional molecularly targeted drugs for liver cancer show some problems, such as short residence time, poor drug enrichment, and drug resistance. Therefore, developing new diagnosis and treatment methods to effectively improve the diagnosis, treatment, and long-term prognosis of liver cancer is urgent. As an emerging discipline, nanobiotechnology, based on safe, stable, and efficient nanomaterials, constructs highly targeted nanocarriers according to the unique characteristics of tumors and further derives a variety of efficient diagnosis and treatment methods based on this transport system, providing a new method for the accurate diagnosis and treatment of liver cancer. This paper aims to summarize the latest progress in this field according to existing research and the latest clinical diagnosis and treatment guidelines in hepatocellular carcinoma (HCC), as well as clarify the role, application limitations, and prospects of research on nanomaterials and the development and application of nanotechnology in the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Mengjiao Xu
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, 8 Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Liu Yang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, 8 Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Yanjie Lin
- Department of Hepatology Division 2, Peking University Ditan Teaching Hospital, 8 Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Yao Lu
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, 8 Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Xiaoyue Bi
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, 8 Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Tingting Jiang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, 8 Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Wen Deng
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, 8 Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Lu Zhang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, 8 Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Wei Yi
- Department of Gynecology and Obstetrics, Beijing Ditan Hospital, Capital Medical University, 8 Jingshun East Street, Chaoyang District, Beijing, 100015, China.
| | - Yao Xie
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, 8 Jingshun East Street, Chaoyang District, Beijing, 100015, China.
- Department of Hepatology Division 2, Peking University Ditan Teaching Hospital, 8 Jingshun East Street, Chaoyang District, Beijing, 100015, China.
| | - Minghui Li
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, 8 Jingshun East Street, Chaoyang District, Beijing, 100015, China.
- Department of Hepatology Division 2, Peking University Ditan Teaching Hospital, 8 Jingshun East Street, Chaoyang District, Beijing, 100015, China.
| |
Collapse
|
6
|
Structural and Functional Basis of JAMM Deubiquitinating Enzymes in Disease. Biomolecules 2022; 12:biom12070910. [PMID: 35883466 PMCID: PMC9313428 DOI: 10.3390/biom12070910] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 02/04/2023] Open
Abstract
Deubiquitinating enzymes (DUBs) are a group of proteases that are important for maintaining cell homeostasis by regulating the balance between ubiquitination and deubiquitination. As the only known metalloproteinase family of DUBs, JAB1/MPN/Mov34 metalloenzymes (JAMMs) are specifically associated with tumorigenesis and immunological and inflammatory diseases at multiple levels. The far smaller numbers and distinct catalytic mechanism of JAMMs render them attractive drug targets. Currently, several JAMM inhibitors have been successfully developed and have shown promising therapeutic efficacy. To gain greater insight into JAMMs, in this review, we focus on several key proteins in this family, including AMSH, AMSH-LP, BRCC36, Rpn11, and CSN5, and emphatically discuss their structural basis, diverse functions, catalytic mechanism, and current reported inhibitors targeting JAMMs. These advances set the stage for the exploitation of JAMMs as a target for the treatment of various diseases.
Collapse
|
7
|
Integrin α2 and β1 Cross-Communication with mTOR/AKT and the CDK-Cyclin Axis in Hepatocellular Carcinoma Cells. Cancers (Basel) 2022; 14:cancers14102430. [PMID: 35626034 PMCID: PMC9139686 DOI: 10.3390/cancers14102430] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/10/2022] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Hepatocellular carcinoma (HCC) progression depends on two major processes, tumor growth and invasion. The present study investigated how these events are linked. A panel of HCC cell lines were stimulated with insulin-like growth factor-1 (IGF1) and the biological behavior was evaluated. IGF1 activated the proliferation and invasion cascade by altering the expression level of integrin α subtypes, which were associated with the AKT-mTOR pathway and the CDK-Cyclin axis. We assume that HCC progression is controlled by a fine-tuned network between IGF1 driven integrin signaling, the Akt-mTOR pathway, and the CDK-Cyclin axis. Concerted targeting of these pathways may, therefore, become an innovative option to prevent cancer dissemination. Abstract Integrin receptors contribute to hepatocellular carcinoma (HCC) invasion, while AKT-mTOR signaling controls mitosis. The present study was designed to explore the links between integrins and the AKT-mTOR pathway and the CDK-Cyclin axis. HCC cell lines (HepG2, Huh7, Hep3B) were stimulated with soluble collagen or Matrigel to activate integrins, or with insulin-like growth factor 1 (IGF1) to activate AKT-mTOR. HCC growth, proliferation, adhesion, and chemotaxis were evaluated. AKT/mTOR-related proteins, proteins of the CDK-Cyclin axis, focal adhesion kinase (FAK), and integrin-linked kinase (ILK) were determined following IGF1-stimulation or integrin knockdown. Stimulation with collagen or Matrigel increased tumor cell growth and proliferation. This was associated with significant alteration of the integrins α2, αV, and β1. Blockade of these integrins led to cell cycle arrest in G2/M and diminished the number of tumor cell clones. Knocking down the integrins α2 or β1 suppressed ILK, reduced FAK-phosphorylation and diminished AKT/mTOR, as well as the proteins of the CDK-Cyclin axis. Activating the cells with IGF1 enhanced the expression of the integrins α2, αV, β1, activated FAK, and increased tumor cell adhesion and chemotaxis. Blocking the AKT pathway canceled the enhancing effect of IGF on the integrins α2 and β1. These findings reveal that HCC growth, proliferation, and invasion are controlled by a fine-tuned network between α2/β1-FAK signaling, the AKT-mTOR pathway, and the CDK–Cyclin axis. Concerted blockade of the integrin α2/β1 complex along with AKT-mTOR signaling could, therefore, provide an option to prevent progressive dissemination of HCC.
Collapse
|
8
|
Lei H, Wang J, Hu J, Zhu Q, Wu Y. Deubiquitinases in hematological malignancies. Biomark Res 2021; 9:66. [PMID: 34454635 PMCID: PMC8401176 DOI: 10.1186/s40364-021-00320-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022] Open
Abstract
Deubiquitinases (DUBs) are enzymes that control the stability, interactions or localization of most cellular proteins by removing their ubiquitin modification. In recent years, some DUBs, such as USP7, USP9X and USP10, have been identified as promising therapeutic targets in hematological malignancies. Importantly, some potent inhibitors targeting the oncogenic DUBs have been developed, showing promising inhibitory efficacy in preclinical models, and some have even undergone clinical trials. Different DUBs perform distinct function in diverse hematological malignancies, such as oncogenic, tumor suppressor or context-dependent effects. Therefore, exploring the biological roles of DUBs and their downstream effectors will provide new insights and therapeutic targets for the occurrence and development of hematological malignancies. We summarize the DUBs involved in different categories of hematological malignancies including leukemia, multiple myeloma and lymphoma. We also present the recent development of DUB inhibitors and their applications in hematological malignancies. Together, we demonstrate DUBs as potential therapeutic drug targets in hematological malignancies.
Collapse
Affiliation(s)
- Hu Lei
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jiaqi Wang
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiacheng Hu
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qian Zhu
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yingli Wu
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
9
|
Identification of a seven-long non-coding RNA signature associated with Jab1/CSN5 in predicting hepatocellular carcinoma. Cell Death Discov 2021; 7:178. [PMID: 34247194 PMCID: PMC8272716 DOI: 10.1038/s41420-021-00560-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/10/2021] [Accepted: 06/13/2021] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer death worldwide, accounting for over 700,000 deaths each year. The lack of predictive and prognostic biomarkers for HCC, with effective therapy, remains a significant challenge for HCC management. Long non-coding RNAs (lncRNAs) play a key role in tumorigenesis and have clinical value as potential biomarkers in the early diagnosis and prediction of HCC. Jun activation domain-binding protein 1 (Jab1, also known as COP9 signalosome subunit 5, CSN5) is a potential oncogene that plays a critical role in the occurrence of HCC. Here, we performed a comprehensive analysis for Jab1/CSN5-associated lncRNAs to predict the prognosis of HCC. The differentially expressed (DE) lncRNAs between in HCC were analyzed based on the TCGA RNA-seq data. We detected 1031 upregulated lncRNAs in 371 HCC tissues and identified a seven-lncRNA signature strongly correlated with Jab1/CSN5 (SNHG6, CTD3065J16.9, LINC01604, CTD3025N20.3, KB-1460A1.5, RP13-582O9.7, and RP11-29520.2). We further evaluated the prognostic significance of these lncRNAs by GEPIA (http://gepia.cancer-pku.cn/). The expression data in 364 liver tumors indicated that this seven-lncRNA signature could better predict worse survival in HCC patients. Moreover, 35 clinical HCC samples were evaluated to assess the validity and reproducibility of the bioinformatic analysis. We found that the targeted lncRNAs were upregulated, with a strong association with Jab1/CSN5 and prognostic value in HCC. Functional enrichment analysis by Gene Ontology (GO) showed that these seven prognostic lncRNAs exhibit oncogenic properties and are associated with prominent hallmarks of cancer. Overall, our findings demonstrate the clinical implication of Jab1/CSN5 with the seven‐lncRNAs in predicting survival for patients with HCC.
Collapse
|
10
|
Ruan Z, Liang M, Shang L, Lai M, Deng X, Su X. Shikonin-mediated PD-L1 degradation suppresses immune evasion in pancreatic cancer by inhibiting NF-κB/STAT3 and NF-κB/CSN5 signaling pathways. Pancreatology 2021; 21:630-641. [PMID: 33707115 DOI: 10.1016/j.pan.2021.01.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pancreatic cancer (PC) is a highly fatal malignancy with few effective therapies currently available. Recent studies have shown that PD-L1 inhibitors could be potential therapeutic targets for the treatment of PC. The present study aims to investigate the effect of Shikonin on immune evasion in PC with the involvement of the PD-L1 degradation. METHODS Initially, the expression patterns of PD-L1 and NF-κB in PC were predicted in-silico using the GEPIA database, and were subsequently validated using PC tissues. Thereafter, the correlation of NF-κB with STAT3, CSN5 and PD-L1 was examined. PC cells were treated with Shikonin, NF-κB inhibitor, STAT3 activator, and CSN5 overexpression plasmid to investigate effects on PD-L1 glycosylation and immune evasion in PC. Finally, in vivo tumor formation was induced in C57BL/6J mice, in order to verify the in vitro results. RESULTS PD-L1, NF-κB, NF-κB p65, STAT3, and CSN5 were highly expressed in PC samples, and NF-κB was positively correlated with STAT3/CSN5/PD-L1. Inhibition of NF-κB decreased PD-L1 glycosylation and increased PD-L1 degradation, whereas activated STAT3 and overexpressed CSN5 reversed these trends. Shikonin blocked immune evasion in PC, and lowered the expression of PD-L1, NF-κB, NF-κB p65, STAT3 and CSN5 in vivo and in vitro. CONCLUSION The findings indicated Shikonin inhibited immune evasion in PC by inhibiting PD-L1 glycosylation and activating the NF-κB/STAT3 and NF-κB/CSN5 signaling pathways. These effects of Shikonin on PC cells may bear important potential therapeutic implications for the treatment of PC.
Collapse
Affiliation(s)
- Zhiyan Ruan
- School of Pharmacy, Guangdong Food & Drug Vocational College, Guangzhou, 510520, PR China
| | - Minhua Liang
- School of Pharmacy, Guangdong Food & Drug Vocational College, Guangzhou, 510520, PR China
| | - Ling Shang
- School of Pharmacy, Guangdong Food & Drug Vocational College, Guangzhou, 510520, PR China
| | - Manxiang Lai
- School of Pharmacy, Guangdong Food & Drug Vocational College, Guangzhou, 510520, PR China
| | - Xiangliang Deng
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Xinguo Su
- School of Pharmacy, Guangdong Food & Drug Vocational College, Guangzhou, 510520, PR China.
| |
Collapse
|
11
|
Xie P, Wang H, Fang J, Du D, Tian Z, Zhen J, Liu Y, Ding Y, Fu B, Liu F, Huang D, Yu J. CSN5 Promotes Carcinogenesis of Thyroid Carcinoma Cells Through ANGPTL2. Endocrinology 2021; 162:6122687. [PMID: 33508120 DOI: 10.1210/endocr/bqaa206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Indexed: 12/13/2022]
Abstract
COP9 signalosome subunit 5 (CSN5) plays a key role in carcinogenesis of multiple cancers and contributes to the stabilization of target proteins through deubiquitylation. However, the underlying role of CSN5 in thyroid carcinoma has not been reported. In this research, our data showed that CSN5 was overexpressed in thyroid carcinoma tissues compared with paracancerous tissues. Furthermore, a series of gain/loss functional assays were performed to demonstrate the role of CSN5 in facilitating thyroid carcinoma cell proliferation and metastasis. Additionally, we found there was a positive correlation between CSN5 and angiopoietin-like protein 2 (ANGPTL2) protein levels in thyroid carcinoma tissues and that CSN5 promoted thyroid carcinoma cell proliferation and metastasis through ANGPTL2. We also identified the underlying mechanism that CSN5 elevated ANGPTL2 protein level by directly binding it, decreasing its ubiquitination and degradation. Overall, our results highlight the significance of CSN5 in promoting thyroid carcinoma carcinogenesis and implicate CSN5 as a promising candidate for thyroid carcinoma treatment.
Collapse
Affiliation(s)
- Peiyi Xie
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hui Wang
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, China National Research Center for Metabolic Diseases, Shanghai Jiao Tong University School of Medicine, China
| | - Jiayu Fang
- Second College of Clinical Medicine, Nanchang University, China
| | - Dongnian Du
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ze Tian
- Second College of Clinical Medicine, Nanchang University, China
| | - Jing Zhen
- Second College of Clinical Medicine, Nanchang University, China
| | - Yue Liu
- Second College of Clinical Medicine, Nanchang University, China
| | - Yongqi Ding
- Second College of Clinical Medicine, Nanchang University, China
| | - Bidong Fu
- Second College of Clinical Medicine, Nanchang University, China
| | - Fanrong Liu
- Department of Pathology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Da Huang
- Department of Thyroid Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Jichun Yu
- Department of Thyroid Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
12
|
A system biological approach to investigate the genetic profiling and comorbidities of type 2 diabetes. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100830] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
13
|
Berkel C, Cacan E. DYNLL1 is hypomethylated and upregulated in a tumor stage- and grade-dependent manner and associated with increased mortality in hepatocellular carcinoma. Exp Mol Pathol 2020; 117:104567. [PMID: 33171156 DOI: 10.1016/j.yexmp.2020.104567] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/23/2020] [Accepted: 11/03/2020] [Indexed: 01/22/2023]
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide and cellular mechanisms regulating HCC pathogenesis and progression are not completely understood. DYNLL1 is essential for the development and expansion of MYC-driven B cell lymphoma, and also regulates genomic stability and responses to DNA-damaging chemotherapy in BRCA1-deficient tumors. However, the role and regulation of DYNLL1 has not been previously studied in the context of HCC. Here we report that DYNLL1 gene is hypomethylated and its expression is upregulated in HCC patients compared to healthy controls. The expression of DYNLL1 changes in a tumor grade- and stage-dependent manner in HCC. In this study, we further show that high DYNLL1 expression results in shorter overall and progression-free survival in hepatocellular carcinoma patients. Similar to DYNLL1, one of its protein interactors, RACK1, also shows decreased CpG-aggregated methylation and increased expression in HCC. RACK1 expression increases from early to late stage and from low to high grade in HCC. We found that high RACK1 expression is significantly associated with increased mortality of HCC patients. The present study shows that the epigenetic regulation of DYNLL1 and its consequent upregulation might be contributing to cancer development and progression in HCC. Its higher expression in late stage or high grade HCC may favor more aggressive disease as pointed by the increased mortality in high expression cohort. A better mechanistic understanding of the role of DYNLL1 in HCC will be needed to develop targeted treatment strategies in the future.
Collapse
Affiliation(s)
- Caglar Berkel
- Department of Molecular Biology and Genetics, Tokat Gaziosmanpasa University, Tokat, Turkey.
| | - Ercan Cacan
- Department of Molecular Biology and Genetics, Tokat Gaziosmanpasa University, Tokat, Turkey.
| |
Collapse
|
14
|
Abstract
Klebsiella pneumoniae has been singled out as an urgent threat to human health due to the increasing isolation of strains resistant to “last-line” antimicrobials, narrowing the treatment options against Klebsiella infections. Unfortunately, at present, we cannot identify candidate compounds in late-stage development for treatment of multidrug-resistant Klebsiella infections; this pathogen is exemplary of the mismatch between unmet medical needs and the current antimicrobial research and development pipeline. Furthermore, there is still limited evidence on K. pneumoniae pathogenesis at the molecular and cellular levels in the context of the interactions between bacterial pathogens and their hosts. In this research, we have uncovered a sophisticated strategy employed by Klebsiella to subvert the activation of immune defenses by controlling the modification of proteins. Our research may open opportunities to develop new therapeutics based on counteracting this Klebsiella-controlled immune evasion strategy. Klebsiella pneumoniae is an important cause of multidrug-resistant infections worldwide. Understanding the virulence mechanisms of K. pneumoniae is a priority and timely to design new therapeutics. Here, we demonstrate that K. pneumoniae limits the SUMOylation of host proteins in epithelial cells and macrophages (mouse and human) to subvert cell innate immunity. Mechanistically, in lung epithelial cells, Klebsiella increases the levels of the deSUMOylase SENP2 in the cytosol by affecting its K48 ubiquitylation and its subsequent degradation by the ubiquitin proteasome. This is dependent on Klebsiella preventing the NEDDylation of the Cullin-1 subunit of the ubiquitin ligase complex E3-SCF-βTrCP by exploiting the CSN5 deNEDDylase. Klebsiella induces the expression of CSN5 in an epidermal growth factor receptor (EGFR)-phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT)-extracellular signal-regulated kinase (ERK)-glycogen synthase kinase 3 beta (GSK3β) signaling pathway-dependent manner. In macrophages, Toll-like receptor 4 (TLR4)-TRAM-TRIF-induced type I interferon (IFN) via IFN receptor 1 (IFNAR1)-controlled signaling mediates Klebsiella-triggered decrease in the levels of SUMOylation via let-7 microRNAs (miRNAs). Our results revealed the crucial role played by Klebsiella polysaccharides, the capsule, and the lipopolysaccharide (LPS) O-polysaccharide, to decrease the levels of SUMO-conjugated proteins in epithelial cells and macrophages. A Klebsiella-induced decrease in SUMOylation promotes infection by limiting the activation of inflammatory responses and increasing intracellular survival in macrophages.
Collapse
|
15
|
Guo D, Zhang D, Ren M, Lu G, Zhang X, He S, Li Y. THBS4 promotes HCC progression by regulating ITGB1 via FAK/PI3K/AKT pathway. FASEB J 2020; 34:10668-10681. [PMID: 32567740 DOI: 10.1096/fj.202000043r] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/07/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022]
Abstract
Our research aims to identify the role of thrombospondin 4 (THBS4) in hepatocellular carcinoma (HCC). First, bioinformatic analysis was applied for detection the expression of THBS4 in HCC samples, and qRT-PCR and western blot were performed to explore the expression of THBS4 in HCC tissues and adjacent samples. Next, colony formation assay and cell viability assay were used to assess the function of THBS4 on HCC cells growth while transwell assay and scratch test were for metastasis. Meanwhile Xenograft tumor models were further conducted to verify the function of THBS4 in HCC. As for mechanism in deep, we investigated the influence of THBS4 on epithelial-mesenchymal transition (EMT) development and the interaction between THBS4 and integrin (ITG) family using multiple experiments, including western blot, immunofluorescence and immunoprecipitation (IP). As a result, our research discovered that the overexpression of THBS4 in both HCC patients' tissues and cell lines mediates HCC cells proliferation and metastasis in vitro and in vivo. In-depth, THBS4 regulated EMT progression and interacted with ITG family to modulate FAK/PI3K/AKT pathway. In conclusion, THBS4 as an oncogene interacts with integrinβ1 (ITGB1) to regulate HCC development via FAK/PI3K/AKT pathway.
Collapse
Affiliation(s)
- Dan Guo
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Dan Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Mudan Ren
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Guifang Lu
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xu Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Shuixiang He
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Yarui Li
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
16
|
The crucial p53-dependent oncogenic role of JAB1 in osteosarcoma in vivo. Oncogene 2020; 39:4581-4591. [PMID: 32390003 PMCID: PMC7274902 DOI: 10.1038/s41388-020-1320-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 04/23/2020] [Accepted: 04/28/2020] [Indexed: 12/25/2022]
Abstract
Osteosarcoma (OS) is the most common primary bone cancer and ranks amongst the leading causes of cancer mortality in young adults. Jun activation domain binding protein 1 (JAB1) is overexpressed in many cancers and has recently emerged as a novel target for cancer treatment. However, the role of JAB1 in osteosarcoma was virtually unknown. In this study, we demonstrate that JAB1-knockdown in malignant osteosarcoma cell lines significantly reduced their oncogenic properties, including proliferation, colony formation, and motility. We also performed RNA-sequencing analysis in JAB1-knockdown OS cells and identified 4110 genes that are significantly differentially expressed. This demonstrated for the first time that JAB1 regulates a large and specific transcriptome in cancer. We also found that JAB1 is overexpressed in human OS and correlates with a poor prognosis. Moreover, we generated a novel mouse model that overexpresses Jab1 specifically in osteoblasts upon a TP53 heterozygous sensitizing background. Interestingly, by 13 months of age, a significant proportion of these mice spontaneously developed conventional OS. Finally, we demonstrate that a novel, highly specific small molecule inhibitor of JAB1, CSN5i-3, reduces osteosarcoma cell viability and has specific effects on the ubiquitin-proteasome system in OS. Thus, we show for the first time that the overexpression of JAB1 in vivo can result in accelerated spontaneous tumor formation in a p53-dependent manner. In summary, JAB1 might be a unique target for the treatment of osteosarcoma and other cancers.
Collapse
|
17
|
Structural dynamics of the human COP9 signalosome revealed by cross-linking mass spectrometry and integrative modeling. Proc Natl Acad Sci U S A 2020; 117:4088-4098. [PMID: 32034103 DOI: 10.1073/pnas.1915542117] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The COP9 signalosome (CSN) is an evolutionarily conserved eight-subunit (CSN1-8) protein complex that controls protein ubiquitination by deneddylating Cullin-RING E3 ligases (CRLs). The activation and function of CSN hinges on its structural dynamics, which has been challenging to decipher by conventional tools. Here, we have developed a multichemistry cross-linking mass spectrometry approach enabled by three mass spectometry-cleavable cross-linkers to generate highly reliable cross-link data. We applied this approach with integrative structure modeling to determine the interaction and structural dynamics of CSN with the recently discovered ninth subunit, CSN9, in solution. Our results determined the localization of CSN9 binding sites and revealed CSN9-dependent structural changes of CSN. Together with biochemical analysis, we propose a structural model in which CSN9 binding triggers CSN to adopt a configuration that facilitates CSN-CRL interactions, thereby augmenting CSN deneddylase activity. Our integrative structure analysis workflow can be generalized to define in-solution architectures of dynamic protein complexes that remain inaccessible to other approaches.
Collapse
|
18
|
Parashar D, Rajendran V, Shukla R, Sistla R. Lipid-based nanocarriers for delivery of small interfering RNA for therapeutic use. Eur J Pharm Sci 2020; 142:105159. [DOI: 10.1016/j.ejps.2019.105159] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/03/2019] [Accepted: 11/15/2019] [Indexed: 12/14/2022]
|
19
|
Chemogenomic study of gemcitabine using Saccharomyces cerevisiae as model cell-molecular insights about chemoresistance. Braz J Microbiol 2019; 51:489-496. [PMID: 31515725 DOI: 10.1007/s42770-019-00154-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/06/2019] [Indexed: 12/14/2022] Open
Abstract
Gemcitabine (GEM) is the drug used as first line to treat pancreatic cancer, one of the most devastating human tumors. This peculiar type of tumor develops resistance to several drugs, including GEM, due to its desmoplastic reaction and other features. The GEM chemoresistance has been investigated at molecular level aiming to find a pathway whose inhibition or activation should overcome it. Through next-generation sequencing was performed a chemogenomic assay of GEM using Saccharomyces cerevisiae as model cell and the results showed that more than 40% of genes related to GEM response in yeast possess unknown or dubious function. We choose two yeast mutants to individually validate the fitness defect results observed by chemogenomic assay, Δhmt1 and Δcsi1, and it was found that in addition to some already described pathways involved in GEM resistance, cells deficient in deneddylation enzyme Cop9 Signalosome Interactor 1 (Csi1p) presented a high sensitivity to GEM. This was confirmed by individual growth analyses of Δcsi1 cells exposed to GEM, and this phenotype was reverted with CSI1 complementation gene. Csi1p is a well-characterized homolog equivalent to human Csn6 subunit of COP9 signalosome (CSN) involved in deneddylation process. We highlighted too that epigenetic alterations, such as methylation mediated by protein arginine methyltransferase 1, play an important role in regulating gemcitabine treatment resistance. Our results point out new unexplored molecular pathways that can be used to overcome GEM resistance: the inhibition of CSN and the arginine methyltransferase activities.
Collapse
|
20
|
CSNAP, the smallest CSN subunit, modulates proteostasis through cullin-RING ubiquitin ligases. Cell Death Differ 2019; 27:984-998. [PMID: 31367012 DOI: 10.1038/s41418-019-0392-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 11/09/2022] Open
Abstract
The cullin-RING ubiquitin E3 ligase (CRL) family consists of ~250 complexes that catalyze ubiquitylation of proteins to achieve cellular regulation. All CRLs are inhibited by the COP9 signalosome complex (CSN) through both enzymatic (deneddylation) and nonenzymatic (steric) mechanisms. The relative contribution of these two mechanisms is unclear. Here, we decouple the mechanisms using CSNAP, the recently discovered ninth subunit of the CSN. We find that CSNAP reduces the affinity of CSN toward CRL complexes. Removing CSNAP does not affect deneddylation, but leads to global effects on the CRL, causing altered reproductive capacity, suppressed DNA damage response, and delayed cell cycle progression. Thus, although CSNAP is only 2% of the CSN mass, it plays a critical role in the steric regulation of CRLs by the CSN.
Collapse
|
21
|
Mao L, Le S, Jin X, Liu G, Chen J, Hu J. CSN5 promotes the invasion and metastasis of pancreatic cancer by stabilization of FOXM1. Exp Cell Res 2019; 374:274-281. [PMID: 30352219 DOI: 10.1016/j.yexcr.2018.10.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 10/14/2018] [Accepted: 10/19/2018] [Indexed: 01/10/2023]
|
22
|
Guo Z, Wang Y, Zhao Y, Shu Y, Liu Z, Zhou H, Wang H, Zhang W. The pivotal oncogenic role of Jab1/CSN5 and its therapeutic implications in human cancer. Gene 2018; 687:219-227. [PMID: 30468907 DOI: 10.1016/j.gene.2018.11.061] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/01/2018] [Accepted: 11/19/2018] [Indexed: 01/28/2023]
Abstract
Jab1/CSN5 is a conserved multifunctional protein involved in ubiquitin-mediated protein degradation. Deregulation of Jab1/CSN5 can exert dramatic effects on diverse cellular functions, including DNA repair, cell cycle control, apoptosis, angiogenesis, and signal transduction, all of which are critical for tumor development. Although increasing evidence has demonstrated that Jab1/CSN5 was overexpressed in a variety of human cancers and usually correlated with poor prognosis, little was known about the underlying regulatory principles that coordinated its function. In this review, we highlight recent advances of the oncogenic role of Jab1/CSN5 and its potential as a therapeutic target for anticancer intervention.
Collapse
Affiliation(s)
- Zhen Guo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University and Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Youhong Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University and Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Yu Zhao
- Key Laboratory of Translational Radiation Oncology, Hunan Province, Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, PR China
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD 21201, USA
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University and Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University and Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Hui Wang
- Key Laboratory of Translational Radiation Oncology, Hunan Province, Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, PR China.
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University and Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China.
| |
Collapse
|
23
|
Hajiasgharzadeh K, Somi MH, Shanehbandi D, Mokhtarzadeh A, Baradaran B. Small interfering RNA-mediated gene suppression as a therapeutic intervention in hepatocellular carcinoma. J Cell Physiol 2018; 234:3263-3276. [PMID: 30362510 DOI: 10.1002/jcp.27015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 06/25/2018] [Indexed: 12/14/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the lethal and difficult-to-cure cancers worldwide. Owing to the late diagnosis and drug resistance of malignant hepatocytes, treatment of this cancer by conventional chemotherapy agents is challenging, and researchers are seeking new alternative treatment options to overcome therapy resistance in this neoplasm. RNA interference (RNAi) is a potent and specific approach in targeting gene expression and has emerged as a novel therapeutic tool for many diseases, including cancers. Small interfering RNA (siRNA) is a type of RNAi that is produced intracellularly from exogenous synthetic oligonucleotides and can selectively knock down target gene expression in a sequence-specific manner. Various factors play roles in the initiation and progression of HCC and provide multiple candidate targets for siRNA intervention. In addition, due to the liver's unique architecture and availability of some hepatic siRNA delivery methods, this organ has received much more attention as a target tissue for such oligonucleotide action. Recent advances in designing nanoparticle systems for the in vivo delivery of siRNAs have markedly enhanced the potency of siRNA-mediated gene silencing under clinical development for HCC therapy. The utility of siRNAs as anti-HCC agents is the subject of the current review. siRNA-based gene therapies could be one of the main feasible approaches for HCC therapy in the future.
Collapse
Affiliation(s)
| | - Mohammad Hossein Somi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
24
|
Fu Q, Yang F, Zhao J, Yang X, Xiang T, Huai G, Zhang J, Wei L, Deng S, Yang H. Bioinformatical identification of key pathways and genes in human hepatocellular carcinoma after CSN5 depletion. Cell Signal 2018; 49:79-86. [PMID: 29885455 DOI: 10.1016/j.cellsig.2018.06.002] [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] [Received: 04/21/2018] [Revised: 05/31/2018] [Accepted: 06/05/2018] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer. It has been previously reported that CSN5 depletion is an effective method in human HCC. In the current study, we aimed to uncover gene signatures and key pathways during HCC. Gene expression profiles of GSE26485 were downloaded from GEO database. Totally, 101 differentially expressed genes (DEGs) were up-regulated and 146 ones were down-regulated. Biological processes (BP) and Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) analysis showed that the DEGs were mainly enriched in regulation of cell growth, oxidation-reduction process, mitotic cytokinesis, negative regulation of macroautophagy, endosome organization, lysosome, biosynthesis of antibiotics, small cell lung cancer and glutathione metabolism and so on (P < 0.05). Protein-protein interaction (PPI) network, Kaplan-Meier, log-rank method, western blot, immunohistochemistry and encyclopedia of DNA elements (ENCODE) analysis showed that CSN5 depletion took effects through down-regulation of SMAD5-related pathways which include EXO1, CENPA and NCAPG, resulting in the inactivation of H3K4me3 and H3K36me3. Those genes represent the promising targets for therapeutic intervention in HCC patients.
Collapse
Affiliation(s)
- Qiang Fu
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan province, China; Organ Transplantation translational medicine Key laboratory of Sichuan province,Chengdu, Sichuan 610072, China
| | - Fan Yang
- Women and Children Health Care Center of Luoyang, Luoyang 471000, Henan province, China
| | - Ji Zhao
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan province, China; Organ Transplantation translational medicine Key laboratory of Sichuan province,Chengdu, Sichuan 610072, China
| | - Xingxing Yang
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan province, China; Organ Transplantation translational medicine Key laboratory of Sichuan province,Chengdu, Sichuan 610072, China
| | - Tengxiao Xiang
- People's Hospital of Changshou Chongqing, Chongqing 401220, China
| | - Guoli Huai
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan province, China; Organ Transplantation translational medicine Key laboratory of Sichuan province,Chengdu, Sichuan 610072, China
| | - Jiashu Zhang
- Organ Transplantation translational medicine Key laboratory of Sichuan province,Chengdu, Sichuan 610072, China; North Sichuan Medical College, Nanchong 637100, Sichuan province, China
| | - Liang Wei
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan province, China; Organ Transplantation translational medicine Key laboratory of Sichuan province,Chengdu, Sichuan 610072, China
| | - Shaoping Deng
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan province, China; Organ Transplantation translational medicine Key laboratory of Sichuan province,Chengdu, Sichuan 610072, China; North Sichuan Medical College, Nanchong 637100, Sichuan province, China; Human Islet Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston 02114, MA, USA.
| | - Hongji Yang
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan province, China; Organ Transplantation translational medicine Key laboratory of Sichuan province,Chengdu, Sichuan 610072, China.
| |
Collapse
|
25
|
Liu G, Claret FX, Zhou F, Pan Y. Jab1/COPS5 as a Novel Biomarker for Diagnosis, Prognosis, Therapy Prediction and Therapeutic Tools for Human Cancer. Front Pharmacol 2018; 9:135. [PMID: 29535627 PMCID: PMC5835092 DOI: 10.3389/fphar.2018.00135] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/07/2018] [Indexed: 12/26/2022] Open
Abstract
C-Jun activation domain-binding protein-1 (Jab1) involves in controlling cellular proliferation, cell cycle, apoptosis, affecting a series of pathways, as well as regulating genomic instability and DNA damage response (DDR). Jab1/COPS5 dysregulation contributes to oncogenesis by deactivating several tumor suppressors and activating oncogenes. Jab1 overexpression was found in many tumor types, illuminating its important role in cancer initiation, progression, and prognosis. Jab1/COPS5 has spurred a strong research interest in developing inhibitors of oncogenes/oncoproteins for cancer therapy. In this paper, we present evidences demonstrating the importance of Jab1/COPS5 overexpression in several cancer types and recent advances in dissecting the Jab1/COPS5 upstream and downstream signaling pathways. By conducting ingenuity pathway analysis (IPA) based on the Ingenuity Knowledge Base, we investigated signaling network that interacts with Jab1/COPS5. The data confirmed the important role of Jab1/COPS5 in tumorigenesis, demonstrating the potential of Jab1/COPS5 to be used as a biomarker for cancer patients, and further support that Jab1/COPS5 may serve as a potential therapeutic target in different cancers.
Collapse
Affiliation(s)
- Guohong Liu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Department of Systems Biology, The University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Francois X. Claret
- Department of Systems Biology, The University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yunbao Pan
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
26
|
Li PH, Wang L, Pan YJ, Sang MM, Zheng JN, Pei DS. Suppression of Jab1 expression inhibits proliferation and promotes apoptosis of AMC-HN-8 cells. Oncol Lett 2018; 15:5137-5142. [PMID: 29552148 DOI: 10.3892/ol.2018.7963] [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: 06/06/2016] [Accepted: 10/12/2017] [Indexed: 01/10/2023] Open
Abstract
c-Jun activation domain-binding protein-1 (Jab1) is a multifunctional protein involved in cell proliferation and apoptosis, DNA damage and repair and genome stability. In a number of types of human carcinoma, the abnormal expression of Jab1 is associated with poor prognosis, suggesting that Jab1 serves a vital function in tumorigenesis. However, the functional effects and the underlying molecular mechanisms of Jab1 in laryngeal squamous cell carcinoma (LSCC) progression remain poorly understood. The results of the present study demonstrate that downregulating Jab1 expression promotes LSCC apoptosis while inhibiting the proliferation of LSCC cells. Furthermore, Jab1 inhibition results in decreased protein kinase B phosphorylation accompanied by increased caspase-3 cleavage and p53 expression. It has been identified that the increased expression of Jab1 is markedly associated with LSCC progression, therefore Jab1 may be used as a novel target for the treatment of laryngeal cancer.
Collapse
Affiliation(s)
- Pei-Hua Li
- Department of Otorhinolaryngology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Lin Wang
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Yao-Jie Pan
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Department of Oncology, The Affiliated Yancheng Hospital of Medicine School of Southeast University, Yancheng, Jiangsu 224001, P.R. China
| | - Miao-Miao Sang
- Department of Rehabilitation Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Jun-Nian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Dong-Sheng Pei
- Department of Pathology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
27
|
Milcovich G, Lettieri S, Antunes FE, Medronho B, Fonseca AC, Coelho JFJ, Marizza P, Perrone F, Farra R, Dapas B, Grassi G, Grassi M, Giordani S. Recent advances in smart biotechnology: Hydrogels and nanocarriers for tailored bioactive molecules depot. Adv Colloid Interface Sci 2017; 249:163-180. [PMID: 28527520 DOI: 10.1016/j.cis.2017.05.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/06/2017] [Accepted: 05/08/2017] [Indexed: 12/18/2022]
Abstract
Over the past ten years, the global biopharmaceutical market has remarkably grown, with ten over the top twenty worldwide high performance medical treatment sales being biologics. Thus, biotech R&D (research and development) sector is becoming a key leading branch, with expanding revenues. Biotechnology offers considerable advantages compared to traditional therapeutic approaches, such as reducing side effects, specific treatments, higher patient compliance and therefore more effective treatments leading to lower healthcare costs. Within this sector, smart nanotechnology and colloidal self-assembling systems represent pivotal tools able to modulate the delivery of therapeutics. A comprehensive understanding of the processes involved in the self-assembly of the colloidal structures discussed therein is essential for the development of relevant biomedical applications. In this review we report the most promising and best performing platforms for specific classes of bioactive molecules and related target, spanning from siRNAs, gene/plasmids, proteins/growth factors, small synthetic therapeutics and bioimaging probes.
Collapse
Affiliation(s)
- Gesmi Milcovich
- Nano Carbon Materials Research Lab, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
| | - Stefania Lettieri
- Nano Carbon Materials Research Lab, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
| | - Filipe E Antunes
- Coimbra Chemistry Centre, Dept. of Chemistry, University of Coimbra, Rua Larga, Coimbra, Portugal
| | - Bruno Medronho
- Faculty of Sciences and Technology (MEDITBIO), University of Algarve, Campus de Gambelas, Ed. 8, 8005-139 Faro, Portugal
| | - Ana C Fonseca
- CEMMPRE, Department of Chemical Engineering, University of Coimbra, Polo II, Rua Silvio Lima, Coimbra, Portugal
| | - Jorge F J Coelho
- CEMMPRE, Department of Chemical Engineering, University of Coimbra, Polo II, Rua Silvio Lima, Coimbra, Portugal
| | - Paolo Marizza
- Department of Micro- and Nanotechnology, Technical University of Denmark (DTU), Ørsteds Plads Bygning 345Ø, Kongens Lyngby 2800, Denmark
| | - Francesca Perrone
- Department of Life Sciences, Cattinara Hospital, University of Trieste, Strada di Fiume 447, 34100 Trieste, Italy
| | - Rossella Farra
- Department of Life Sciences, Cattinara Hospital, University of Trieste, Strada di Fiume 447, 34100 Trieste, Italy
| | - Barbara Dapas
- Department of Life Sciences, Cattinara Hospital, University of Trieste, Strada di Fiume 447, 34100 Trieste, Italy
| | - Gabriele Grassi
- Department of Life Sciences, Cattinara Hospital, University of Trieste, Strada di Fiume 447, 34100 Trieste, Italy
| | - Mario Grassi
- Department of Engineering and Architecture, University of Trieste, Via Valerio 6, 34127 Trieste, Italy
| | - Silvia Giordani
- Nano Carbon Materials Research Lab, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy; Chemistry Department, Università di Torino, Via Giuria 7, 10125 Turin, Italy.
| |
Collapse
|
28
|
Zhang S, Hong Z, Chai Y, Liu Z, Du Y, Li Q, Liu Q. CSN5 promotes renal cell carcinoma metastasis and EMT by inhibiting ZEB1 degradation. Biochem Biophys Res Commun 2017; 488:101-108. [DOI: 10.1016/j.bbrc.2017.05.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 05/03/2017] [Indexed: 01/10/2023]
|
29
|
Altmann E, Erbel P, Renatus M, Schaefer M, Schlierf A, Druet A, Kieffer L, Sorge M, Pfister K, Hassiepen U, Jones M, Ruedisser S, Ostermeier D, Martoglio B, Jefferson AB, Quancard J. Azaindoles as Zinc‐Binding Small‐Molecule Inhibitors of the JAMM Protease CSN5. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201608672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Eva Altmann
- Novartis Institutes for BioMedical Research Novartis Campus 4002 Basel Switzerland
| | - Paul Erbel
- Novartis Institutes for BioMedical Research Novartis Campus 4002 Basel Switzerland
| | - Martin Renatus
- Novartis Institutes for BioMedical Research Novartis Campus 4002 Basel Switzerland
| | - Michael Schaefer
- Novartis Institutes for BioMedical Research Novartis Campus 4002 Basel Switzerland
| | - Anita Schlierf
- Novartis Institutes for BioMedical Research Novartis Campus 4002 Basel Switzerland
| | - Adelaide Druet
- Novartis Institutes for BioMedical Research Novartis Campus 4002 Basel Switzerland
| | - Laurence Kieffer
- Novartis Institutes for BioMedical Research Novartis Campus 4002 Basel Switzerland
| | - Mickael Sorge
- Novartis Institutes for BioMedical Research Novartis Campus 4002 Basel Switzerland
| | - Keith Pfister
- Novartis Institutes for BioMedical Research Novartis Campus 4002 Basel Switzerland
- Novartis Institutes for BioMedical Research 4560 Horton Street Emeryville CA 94608-2916 USA
| | - Ulrich Hassiepen
- Novartis Institutes for BioMedical Research Novartis Campus 4002 Basel Switzerland
| | - Matthew Jones
- Novartis Institutes for BioMedical Research Novartis Campus 4002 Basel Switzerland
| | - Simon Ruedisser
- Novartis Institutes for BioMedical Research Novartis Campus 4002 Basel Switzerland
| | - Daniela Ostermeier
- Novartis Institutes for BioMedical Research Novartis Campus 4002 Basel Switzerland
| | - Bruno Martoglio
- Novartis Institutes for BioMedical Research Novartis Campus 4002 Basel Switzerland
- Pharma Research and Early Development Roche Innovation Center Basel 4070 Basel Switzerland
| | - Anne B. Jefferson
- Novartis Institutes for BioMedical Research 4560 Horton Street Emeryville CA 94608-2916 USA
- Centers for Therapeutic Innovation Pfizer Inc. San Francisco CA 94158 USA
| | - Jean Quancard
- Novartis Institutes for BioMedical Research Novartis Campus 4002 Basel Switzerland
| |
Collapse
|
30
|
Li P, Xie L, Gu Y, Li J, Xie J. Roles of Multifunctional COP9 Signalosome Complex in Cell Fate and Implications for Drug Discovery. J Cell Physiol 2017; 232:1246-1253. [PMID: 27869306 DOI: 10.1002/jcp.25696] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/18/2016] [Indexed: 01/24/2023]
Abstract
The eight subunits containing COP9 signalosome (CSN) complex, is highly conserved among eukaryotes. CSN, identified as a negative regulator of photomorphogenesis, has also been demonstrated to be important in proteolysis, cellular signal transduction and cell cycle regulation in various eukaryotic organisms. This review mainly summarizes the roles of CSN in cell cycle regulation, signal transduction and apoptosis, and its potential as diagnostic biomarkers, drug targets for cancer and infectious diseases. J. Cell. Physiol. 232: 1246-1253, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ping Li
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing, China
| | - Longxiang Xie
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing, China
| | - Yinzhong Gu
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing, China
| | - Jiang Li
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing, China
| | - Jianping Xie
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing, China
| |
Collapse
|
31
|
Ganbold T, Gerile G, Xiao H, Baigude H. Efficient in vivo siRNA delivery by stabilized d-peptide-based lipid nanoparticles. RSC Adv 2017. [DOI: 10.1039/c6ra25862j] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A lipid functionalized d-dipeptide has shown remarkable biocompatibility and tissue targeting as well as excellent RNAi delivery efficiency in vivo.
Collapse
Affiliation(s)
- Tsogzolmaa Ganbold
- School of Chemistry & Chemical Engineering
- Inner Mongolia University
- Hohhot
- PR China
| | - Gerile Gerile
- School of Chemistry & Chemical Engineering
- Inner Mongolia University
- Hohhot
- PR China
| | - Hai Xiao
- School of Chemistry & Chemical Engineering
- Inner Mongolia University
- Hohhot
- PR China
| | - Huricha Baigude
- School of Chemistry & Chemical Engineering
- Inner Mongolia University
- Hohhot
- PR China
| |
Collapse
|
32
|
Altmann E, Erbel P, Renatus M, Schaefer M, Schlierf A, Druet A, Kieffer L, Sorge M, Pfister K, Hassiepen U, Jones M, Ruedisser S, Ostermeier D, Martoglio B, Jefferson AB, Quancard J. Azaindoles as Zinc-Binding Small-Molecule Inhibitors of the JAMM Protease CSN5. Angew Chem Int Ed Engl 2016; 56:1294-1297. [PMID: 27981705 DOI: 10.1002/anie.201608672] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/27/2016] [Indexed: 11/06/2022]
Abstract
CSN5 is the zinc metalloprotease subunit of the COP9 signalosome (CSN), which is an important regulator of cullin-RING E3 ubiquitin ligases (CRLs). CSN5 is responsible for the cleavage of NEDD8 from CRLs, and blocking deconjugation of NEDD8 traps the CRLs in a hyperactive state, thereby leading to auto-ubiquitination and ultimately degradation of the substrate recognition subunits. Herein, we describe the discovery of azaindoles as a new class of CSN5 inhibitors, which interact with the active-site zinc ion of CSN5 through an unprecedented binding mode. The best compounds inhibited CSN5 with nanomolar potency, led to degradation of the substrate recognition subunit Skp2 in cells, and reduced the viability of HCT116 cells.
Collapse
Affiliation(s)
- Eva Altmann
- Novartis Institutes for BioMedical Research, Novartis Campus, 4002, Basel, Switzerland
| | - Paul Erbel
- Novartis Institutes for BioMedical Research, Novartis Campus, 4002, Basel, Switzerland
| | - Martin Renatus
- Novartis Institutes for BioMedical Research, Novartis Campus, 4002, Basel, Switzerland
| | - Michael Schaefer
- Novartis Institutes for BioMedical Research, Novartis Campus, 4002, Basel, Switzerland
| | - Anita Schlierf
- Novartis Institutes for BioMedical Research, Novartis Campus, 4002, Basel, Switzerland
| | - Adelaide Druet
- Novartis Institutes for BioMedical Research, Novartis Campus, 4002, Basel, Switzerland
| | - Laurence Kieffer
- Novartis Institutes for BioMedical Research, Novartis Campus, 4002, Basel, Switzerland
| | - Mickael Sorge
- Novartis Institutes for BioMedical Research, Novartis Campus, 4002, Basel, Switzerland
| | - Keith Pfister
- Novartis Institutes for BioMedical Research, Novartis Campus, 4002, Basel, Switzerland.,Novartis Institutes for BioMedical Research, 4560 Horton Street, Emeryville, CA, 94608-2916, USA
| | - Ulrich Hassiepen
- Novartis Institutes for BioMedical Research, Novartis Campus, 4002, Basel, Switzerland
| | - Matthew Jones
- Novartis Institutes for BioMedical Research, Novartis Campus, 4002, Basel, Switzerland
| | - Simon Ruedisser
- Novartis Institutes for BioMedical Research, Novartis Campus, 4002, Basel, Switzerland
| | - Daniela Ostermeier
- Novartis Institutes for BioMedical Research, Novartis Campus, 4002, Basel, Switzerland
| | - Bruno Martoglio
- Novartis Institutes for BioMedical Research, Novartis Campus, 4002, Basel, Switzerland.,Pharma Research and Early Development, Roche Innovation Center Basel, 4070, Basel, Switzerland
| | - Anne B Jefferson
- Novartis Institutes for BioMedical Research, 4560 Horton Street, Emeryville, CA, 94608-2916, USA.,Centers for Therapeutic Innovation, Pfizer Inc., San Francisco, CA, 94158, USA
| | - Jean Quancard
- Novartis Institutes for BioMedical Research, Novartis Campus, 4002, Basel, Switzerland
| |
Collapse
|
33
|
Wang S, Pan Y, Zhang R, Xu T, Wu W, Wang C, Huang H, Calin GA, Yang H, Claret FX. Hsa-miR-24-3p increases nasopharyngeal carcinoma radiosensitivity by targeting both the 3'UTR and 5'UTR of Jab1/CSN5. Oncogene 2016; 35:6096-6108. [PMID: 27157611 PMCID: PMC5102828 DOI: 10.1038/onc.2016.147] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 03/11/2016] [Accepted: 03/29/2016] [Indexed: 12/18/2022]
Abstract
Radiotherapy is the standard therapy for nasopharyngeal carcinoma (NPC); however, radioresistance can hinder successful treatment. Here we report that microRNA (miR)-24 acts as a tumor suppressor and radiosensitizer in NPC cells and xenografts by targeting Jab1/CSN5. Although accumulating evidence has shown that Jab1/CSN5 functions as an oncoprotein in human cancers, its regulation through miRs has not been described. In this study, we found that Jab1/CSN5 functioned in a manner opposite to that of miR-24 in NPC tumorigenesis and radioresistance. We demonstrated that miR-24 inhibits Jab1/CSN5 translation via direct binding to its 3' untranslated region (3'UTR) and 5'UTR, leading to tumor growth inhibition, and sensitizes NPC tumors to radiation in vivo. Furthermore, silencing Jab1/CSN5 phenocopied the function of miR-24 in NPC cells after ionizing radiation treatment, resulting in increased apoptosis. Finally, we analyzed 50 paired samples of primary and matched recurrent NPC tissues from 25 NPC patients and subjected them to high-throughput genomic quantitative nuclease protection assay for quantifying simultaneously miR and mRNA levels. Our results showed that miR-24 levels were significantly decreased in recurrent NPC and that levels of Jab1/CSN5, as its target, were higher than those in primary NPC. Together, our findings indicate that miR-24 inhibits NPC tumor growth and increases NPC radiosensitivity by directly regulating Jab1/CSN5 and that both miR-24 and Jab1/CSN5 can serve as prognostic markers for NPC recurrence; this, in turn, may provide a promising therapeutic strategy for reversing NPC radioresistance.
Collapse
Affiliation(s)
- Sumei Wang
- Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510370, P.R. China
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yunbao Pan
- Department of Pathology, affiliated Hospital, Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Rong Zhang
- Department of Endoscopy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Tao Xu
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
- Department of Radiation Oncology, First People’s Hospital of Foshan, Affiliated with Sun Yat-Sen University, Foshan, Guangdong 528000, P.R. China
| | - Wanyin Wu
- Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510370, P.R. China
| | - Chunhua Wang
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Hecheng Huang
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
- Department of Radiation Oncology, Cancer Center, Shantou Central Hospital, Shantou, Guangdong 515000, P.R. China
| | - George A. Calin
- Departments of Experimental Therapeutics and Leukemia and The Center for RNA interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Huiling Yang
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Francois X. Claret
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Experimental Therapeutics Academic Program and Cancer Biology Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| |
Collapse
|
34
|
Schlierf A, Altmann E, Quancard J, Jefferson AB, Assenberg R, Renatus M, Jones M, Hassiepen U, Schaefer M, Kiffe M, Weiss A, Wiesmann C, Sedrani R, Eder J, Martoglio B. Targeted inhibition of the COP9 signalosome for treatment of cancer. Nat Commun 2016; 7:13166. [PMID: 27774986 PMCID: PMC5078989 DOI: 10.1038/ncomms13166] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 09/05/2016] [Indexed: 02/07/2023] Open
Abstract
The COP9 signalosome (CSN) is a central component of the activation and remodelling cycle of cullin-RING E3 ubiquitin ligases (CRLs), the largest enzyme family of the ubiquitin-proteasome system in humans. CRLs are implicated in the regulation of numerous cellular processes, including cell cycle progression and apoptosis, and aberrant CRL activity is frequently associated with cancer. Remodelling of CRLs is initiated by CSN-catalysed cleavage of the ubiquitin-like activator NEDD8 from CRLs. Here we describe CSN5i-3, a potent, selective and orally available inhibitor of CSN5, the proteolytic subunit of CSN. The compound traps CRLs in the neddylated state, which leads to inactivation of a subset of CRLs by inducing degradation of their substrate recognition module. CSN5i-3 differentially affects the viability of tumour cell lines and suppresses growth of a human xenograft in mice. Our results provide insights into how CSN regulates CRLs and suggest that CSN5 inhibition has potential for anti-tumour therapy.
Collapse
Affiliation(s)
- Anita Schlierf
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Eva Altmann
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Jean Quancard
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Anne B Jefferson
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - René Assenberg
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Martin Renatus
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Matthew Jones
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Ulrich Hassiepen
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Michael Schaefer
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Michael Kiffe
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Andreas Weiss
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Christian Wiesmann
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Richard Sedrani
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Jörg Eder
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Bruno Martoglio
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| |
Collapse
|
35
|
Lee SJ, Kim MJ, Kwon IC, Roberts TM. Delivery strategies and potential targets for siRNA in major cancer types. Adv Drug Deliv Rev 2016; 104:2-15. [PMID: 27259398 DOI: 10.1016/j.addr.2016.05.010] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 02/24/2016] [Accepted: 05/15/2016] [Indexed: 02/08/2023]
Abstract
Small interfering RNA (siRNA) has gained attention as a potential therapeutic reagent due to its ability to inhibit specific genes in many genetic diseases. For many years, studies of siRNA have progressively advanced toward novel treatment strategies against cancer. Cancer is caused by various mutations in hundreds of genes including both proto-oncogenes and tumor suppressor genes. In order to develop siRNAs as therapeutic agents for cancer treatment, delivery strategies for siRNA must be carefully designed and potential gene targets carefully selected for optimal anti-cancer effects. In this review, various modifications and delivery strategies for siRNA delivery are discussed. In addition, we present current thinking on target gene selection in major tumor types.
Collapse
|
36
|
Choi Y, Oh ST, Won MA, Choi KM, Ko MJ, Seo D, Jeon TW, Baik IH, Ye SK, Park KU, Park IC, Jang BC, Seo JY, Lee YH. Targeting ODC1 inhibits tumor growth through reduction of lipid metabolism in human hepatocellular carcinoma. Biochem Biophys Res Commun 2016; 478:1674-81. [DOI: 10.1016/j.bbrc.2016.09.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 09/01/2016] [Indexed: 11/29/2022]
|
37
|
Wang L, Zheng JN, Pei DS. The emerging roles of Jab1/CSN5 in cancer. Med Oncol 2016; 33:90. [DOI: 10.1007/s12032-016-0805-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 07/04/2016] [Indexed: 12/13/2022]
|
38
|
Guo H, Jing L, Cheng Y, Atsaves V, Lv Y, Wu T, Su R, Zhang Y, Zhang R, Liu W, Rassidakis GZ, Wei Y, Nan K, Claret FX. Down-regulation of the cyclin-dependent kinase inhibitor p57 is mediated by Jab1/Csn5 in hepatocarcinogenesis. Hepatology 2016; 63:898-913. [PMID: 26606000 DOI: 10.1002/hep.28372] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/23/2015] [Indexed: 01/02/2023]
Abstract
UNLABELLED Down-regulation of p57 (KIP2) cyclin-dependent kinase inhibitors accelerates the growth and invasion of hepatocellular carcinoma (HCC), suggesting that p57 may play an important role in liver carcinogenesis. However, the mechanism or oncogenic signal leading to p57 down-regulation in HCC remains to be determined. Herein, we demonstrated that Jab1/Csn5 expression is negatively correlated with p57 levels in HCC tissues. Kaplan-Meier analysis of tumor samples revealed that high Jab1/Csn5 expression with concurrent low p57 expression is associated with poor overall survival. The inverse pattern of Jab1 and p57 expression was also observed during carcinogenesis in a chemically induced rat HCC model. We also found that mechanistically, Jab1-mediated p57 proteolysis in HCC cells is dependent on 26S-proteasome inhibitors. We further demonstrated that direct physical interaction between Jab1 and p57 triggers p57 down-regulation, independently of Skp2 and Akt pathways, in HCC cells. These data suggest that Jab1 is an important upstream negative regulator of p57 and that aberrant expression of Jab1 in HCC could lead to a significant decrease in p57 levels and contribute to tumor cell growth. Furthermore, restoration of p57 levels induced by loss of Jab1 inhibited tumor cell growth and further increased cell apoptosis in HCC cells. Moreover, silencing Jab1 expression further enhanced the antitumor effects of cisplatin-induced apoptosis in HCC cells. CONCLUSION Jab1-p57 pathway confers resistance to chemotherapy and may represent a potential target for investigational therapy in HCC.
Collapse
Affiliation(s)
- Hui Guo
- Department of Oncology, The First Affiliated Hospital, College of Medicine of Xi'an Jiaotong University, Xi'an, P. R. China.,Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Li Jing
- Department of Oncology, The First Affiliated Hospital, College of Medicine of Xi'an Jiaotong University, Xi'an, P. R. China
| | - Yangzi Cheng
- Department of Oncology, The First Affiliated Hospital, College of Medicine of Xi'an Jiaotong University, Xi'an, P. R. China
| | - Vassilis Atsaves
- Department of Medicine, Division of Critical Care Medicine & Pulmonary Services, University of Athens School of Health Sciences, Athens, Greece
| | - Yi Lv
- Department of Oncology, The First Affiliated Hospital, College of Medicine of Xi'an Jiaotong University, Xi'an, P. R. China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Tao Wu
- Department of Oncology, The First Affiliated Hospital, College of Medicine of Xi'an Jiaotong University, Xi'an, P. R. China
| | - Rujuan Su
- Department of Oncology, The First Affiliated Hospital, College of Medicine of Xi'an Jiaotong University, Xi'an, P. R. China
| | - Yamin Zhang
- Department of Oncology, The First Affiliated Hospital, College of Medicine of Xi'an Jiaotong University, Xi'an, P. R. China
| | - Ronghua Zhang
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Wenbin Liu
- Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - George Z Rassidakis
- Department of Pathology and Cytology, Karolinska University Hospital & Karolinska Institute, Stockholm, Sweden
| | - Yongchang Wei
- Department of Oncology, The First Affiliated Hospital, College of Medicine of Xi'an Jiaotong University, Xi'an, P. R. China
| | - Kejun Nan
- Department of Oncology, The First Affiliated Hospital, College of Medicine of Xi'an Jiaotong University, Xi'an, P. R. China
| | - Francois X Claret
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX.,Experimental Therapeutics Academic Program and Cancer Biology Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX
| |
Collapse
|
39
|
Varshosaz J, Farzan M. Nanoparticles for targeted delivery of therapeutics and small interfering RNAs in hepatocellular carcinoma. World J Gastroenterol 2015; 21:12022-12041. [PMID: 26576089 PMCID: PMC4641122 DOI: 10.3748/wjg.v21.i42.12022] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/31/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the 5th most common malignancy which is responsible for more than half million annual mortalities; also, it is the third leading cause of cancer related death. Unfavorable systemic side-effects of chemotherapeutic agents and susceptibility to the degradation of small interfering RNAs (siRNAs), which can knock down a specific gene involved in the disease, have hampered their clinical application. So, it could be beneficial to develop an efficient carrier for the stabilization and specific delivery of drugs and siRNA to cells. Targeted nanoparticles have gained considerable attention as an efficient drug and gene delivery system, which is due to their capability in achieving the highest accumulation of cytotoxic agents in tumor tissue, modifiable drug pharmacokinetic- and bio-distribution, improved effectiveness of treatment, and limited side-effects. Recent studies have shed more light on the advantages of novel drug loaded carrier systems vs free drugs. Most of the animal studies have reported improvement in treatment efficacy and survival rate using novel carrier systems. Targeted delivery may be achieved passively or actively. In passive targeting, no ligand as homing device is used, while targeting is achieved by incorporating the therapeutic agent into a macromolecule or nanoparticle that passively reaches the target organ. However, in active targeting, the therapeutic agent or carrier system is conjugated to a tissue or cell-specific receptor which is over-expressed in a special malignancy using a ligand called a homing device. This review covers a broad spectrum of targeted nanoparticles as therapeutic and non-viral siRNA delivery systems, which are developed for enhanced cellular uptake and targeted gene silencing in vitro and in vivo and their characteristics and opportunities for the clinical applications of drugs and therapeutic siRNA are discussed in this article. Asialoglycoprotein receptors, low-density lipoprotein, ganglioside GM1 cell surface ligand, epidermal growth factor receptor receptors, monoclonal antibodies, retinoic acid receptors, integrin receptors targeted by Arg-Gly-Asp peptide, folate, and transferrin receptors are the most widely studied cell surface receptors which are used for the site specific delivery of drugs and siRNA-based therapeutics in HCC and discussed in detail in this article.
Collapse
|
40
|
Doan CC, Doan NT, Nguyen QH, Nguyen MH, Do MS, Le VD. Downregulation of Kinesin spindle protein inhibits proliferation, induces apoptosis and increases chemosensitivity in hepatocellular carcinoma cells. IRANIAN BIOMEDICAL JOURNAL 2015; 19:1-16. [PMID: 25605484 PMCID: PMC4322227 DOI: 10.6091/ibj.1386.2014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background: Kinesin spindle protein (KSP) plays a critical role in mitosis. Inhibition of KSP function leads to cell cycle arrest at mitosis and ultimately to cell death. The aim of this study was to suppress KSP expression by specific small-interfering RNA (siRNA) in Hep3B cells and evaluate its anti-tumor activity. Methods: Three siRNA targeting KSP (KSP-siRNA #1-3) and one mismatched-siRNA (Cont-siRNA) were transfected into cells. Subsequently, KSP mRNA and protein levels, cell proliferation, and apoptosis were examined in both Hep3B cells and THLE-3 cells. In addition, the chemosensitivity of KSP-siRNA-treated Hep3B cells with doxorubicin was also investigated using cell proliferation and clonogenic survival assays. Results: The expression of endogenous KSP at both mRNA and protein levels in Hep3B cells was higher than in THLE-3 cells. In Hep3B cells, KSP-siRNA #2 showed a further downregulation of KSP as compared to KSP-siRNA #1 or KSP-siRNA #3. It also exhibited greater suppression of cell proliferation and induction of apoptosis than KSP-siRNA #1 or KSP-siRNA #3; this could be explained by the significant downregulation of cyclin D1, Bcl-2, and survivin. In contrast, KSP-siRNAs had no or lower effects on KSP expression, cell proliferation and apoptosis in THLE-3 cells. We also noticed that KSP-siRNA transfection could increase chemosensitivity to doxorubicin in Hep3B cells, even at low doses compared to control. Conclusion: Reducing the expression level of KSP, combined with drug treatment, yields promising results for eradicating hepatocellular carcinoma (HCC) cells in vitro. This study opens a new direction for liver cancer treatment.
Collapse
Affiliation(s)
- Chinh Chung Doan
- Faculty of Biology, University of Science, Vietnam National University, 227 Nguyen Van Cu Street, Ward 4, District 5, Ho Chi Minh City, Vietnam
| | - Ngoc Trung Doan
- Faculty of Biology, University of Science, Vietnam National University, 227 Nguyen Van Cu Street, Ward 4, District 5, Ho Chi Minh City, Vietnam
| | - Quang Huy Nguyen
- Faculty of Biology, University of Science, Vietnam National University, 227 Nguyen Van Cu Street, Ward 4, District 5, Ho Chi Minh City, Vietnam
| | - Minh Hoa Nguyen
- Faculty of Biology, University of Science, Vietnam National University, 227 Nguyen Van Cu Street, Ward 4, District 5, Ho Chi Minh City, Vietnam
| | - Minh Si Do
- Faculty of Biology, University of Science, Vietnam National University, 227 Nguyen Van Cu Street, Ward 4, District 5, Ho Chi Minh City, Vietnam
| | - Van Dong Le
- Faculty of Biology, University of Science, Vietnam National University, 227 Nguyen Van Cu Street, Ward 4, District 5, Ho Chi Minh City, Vietnam.,Dept. of Immunology, Vietnam Military Medical University,
160 Phung Hung Street, Ha Dong District, Ha Noi City, Vietnam
| |
Collapse
|
41
|
Wang H, Qian Z, Zhao H, Zhang X, Che S, Zhang H, Shang H, Bao J, Hao C, Liu J, Li Z. CSN5 silencing reverses sorafenib resistance of human hepatocellular carcinoma HepG2 cells. Mol Med Rep 2015; 12:3902-3908. [PMID: 26035694 DOI: 10.3892/mmr.2015.3871] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Accepted: 03/18/2015] [Indexed: 01/18/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common tumor types, and is the third leading cause of cancer mortalities worldwide. A large number of patients with HCC are diagnosed at a late stage when the curative treatment of surgical resection and liver transplantation are no longer applicable. Sorafenib has been proved to improve overall survival in advanced HCC; however, drug resistance is common. The present study reported that the CSN5 is correlated with sorafenib resistance of the HCC cell line HepG2/S. Following silencing of CSN5, resistance to sorafenib was reversed, and multi-drug‑resistance proteins, including as adenosine triphosphate binding cassette (ABC)B1, ABCC2 and ABCG2 as well as CDK6, cyclin D1 and B‑cell lymphoma 2 were downregulated. In addition, it was demonstrated that the integrin beta-1, transforming growth factor‑β1 and nuclear factor‑κB pathways were modified by CSN5.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2
- ATP-Binding Cassette Transporters/genetics
- ATP-Binding Cassette Transporters/metabolism
- Antineoplastic Agents/pharmacology
- Apoptosis
- COP9 Signalosome Complex
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Cyclin D1/genetics
- Cyclin D1/metabolism
- Cyclin-Dependent Kinase 6/genetics
- Cyclin-Dependent Kinase 6/metabolism
- Drug Resistance, Neoplasm
- Gene Expression
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
- Hep G2 Cells
- Humans
- Integrin beta1/genetics
- Integrin beta1/metabolism
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Multidrug Resistance-Associated Protein 2
- Multidrug Resistance-Associated Proteins/genetics
- Multidrug Resistance-Associated Proteins/metabolism
- NF-kappa B/physiology
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Niacinamide/analogs & derivatives
- Niacinamide/pharmacology
- Peptide Hydrolases/genetics
- Peptide Hydrolases/metabolism
- Phenylurea Compounds/pharmacology
- RNA, Small Interfering/genetics
- Signal Transduction
- Sorafenib
- Transforming Growth Factor beta1/physiology
Collapse
Affiliation(s)
- Haibo Wang
- Department of Hepatobiliary and Pancreatic Surgery (2), Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Zhengyao Qian
- Department of Cardiology, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Hui Zhao
- Tianjin Entry‑Exit Inspection and Quarantine Bureau, International Travel Healthcare Center, Tianjin 300456, P.R. China
| | - Xibo Zhang
- Department of Hepatobiliary and Pancreatic Surgery (2), Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Shuqiang Che
- Department of Nephrology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300120, P.R. China
| | - Hongtao Zhang
- Department of Hepatobiliary and Pancreatic Surgery (2), Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Haitao Shang
- Department of Hepatobiliary and Pancreatic Surgery (2), Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Jianheng Bao
- Department of Hepatobiliary and Pancreatic Surgery (2), Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Chengfei Hao
- Department of Hepatobiliary and Pancreatic Surgery (2), Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Junjian Liu
- Department of Hepatobiliary and Pancreatic Surgery (2), Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Zhonglian Li
- Department of Hepatobiliary and Pancreatic Surgery (2), Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| |
Collapse
|
42
|
Suppression of CSN5 promotes the apoptosis of gastric cancer cells through regulating p53-related apoptotic pathways. Bioorg Med Chem Lett 2015; 25:2897-901. [DOI: 10.1016/j.bmcl.2015.05.057] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 05/04/2015] [Accepted: 05/22/2015] [Indexed: 02/04/2023]
|
43
|
Martínez T, Jiménez AI, Pañeda C. Short-interference RNAs: becoming medicines. EXCLI JOURNAL 2015; 14:714-46. [PMID: 26648823 PMCID: PMC4669907 DOI: 10.17179/excli2015-297] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 05/12/2015] [Indexed: 12/30/2022]
Abstract
RNA interference is a cellular mechanism by which small molecules of double stranded RNA modulate gene expression acting on the concentration and/or availability of a given messenger RNA. Almost 10 years after Fire and Mello received the Nobel Prize for the discovery of this mechanism in flat worms, RNA interference is on the edge of becoming a new class of therapeutics. With various phase III studies underway, the following years will determine whether RNAi-therapeutics can rise up to the challenge and become mainstream medicines. The present review gives a thorough overview of the current status of this technology focusing on the path to the clinic of this new class of compounds.
Collapse
Affiliation(s)
- Tamara Martínez
- Sylentis, R&D department c/Santiago Grisolía, Tres Cantos, Madrid, Spain
| | - Ana Isabel Jiménez
- Sylentis, R&D department c/Santiago Grisolía, Tres Cantos, Madrid, Spain
| | - Covadonga Pañeda
- Sylentis, R&D department c/Santiago Grisolía, Tres Cantos, Madrid, Spain
| |
Collapse
|
44
|
Pérez-Aguilar B, Vidal CJ, Palomec G, García-Dolores F, Gutiérrez-Ruiz MC, Bucio L, Gómez-Olivares JL, Gómez-Quiroz LE. Acetylcholinesterase is associated with a decrease in cell proliferation of hepatocellular carcinoma cells. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1380-7. [PMID: 25869328 DOI: 10.1016/j.bbadis.2015.04.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 03/27/2015] [Accepted: 04/02/2015] [Indexed: 01/08/2023]
Abstract
Acetylcholinesterase (AChE), the enzyme that rapidly splits acetylcholine into acetate and choline, presents non-cholinergic functions through which may participate in the control of cell proliferation and apoptosis. These two features are relevant in cancer, particularly in hepatocellular carcinoma (HCC), a very aggressive liver tumor with high incidence and poor prognosis in advanced stages. Here we explored the relation between acetylcholinesterase and HCC growth by testing the influence of AChE on proliferation of Huh-7 and HepG2 cell lines, addressed in monolayer cultures, spheroid formation and human liver tumor samples. Results showed a clear relation in AChE expression and cell cycle progression, an effect which depended on cell confluence. Inhibition of AChE activity led to an increase in cell proliferation, which was associated with downregulation of p27 and cyclins. The fact that Huh-7 and HepG2 cell lines provided similar results lent weight to the relationship of AChE expression with cell cycle progression in hepatoma cell lines at least. Human liver tumor samples exhibited a decrease in AChE activity as compared with normal tissue. The evidence presented herein provides additional support for the proposed tumor suppressor role of AChE, which makes it a potential therapeutic target in therapies against hepatocellular carcinoma.
Collapse
Affiliation(s)
- Benjamín Pérez-Aguilar
- Doctorado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, México DF, Mexico; Laboratorio de Fisiología Celular, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, México DF, Mexico
| | - Cecilio J Vidal
- Departamento de Bioquímica y Biología Molecular-A, Universidad de Murcia, IMIB-Arrixaca, Regional Campus of International Excellence "Campus Mare Nostrum", Murcia, Spain
| | - Guillermina Palomec
- Laboratorio de Biomembranas, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, México DF, Mexico
| | | | - María Concepción Gutiérrez-Ruiz
- Laboratorio de Fisiología Celular, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, México DF, Mexico
| | - Leticia Bucio
- Laboratorio de Fisiología Celular, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, México DF, Mexico
| | - José Luis Gómez-Olivares
- Laboratorio de Biomembranas, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, México DF, Mexico.
| | - Luis Enrique Gómez-Quiroz
- Laboratorio de Fisiología Celular, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, México DF, Mexico.
| |
Collapse
|
45
|
Ma X, Yan J, Chen W, Du P, Xie J, Yu H, Wu H. Knockdown of Myosin VI Inhibits Proliferation of Hepatocellular Carcinoma Cells In Vitro. Chem Biol Drug Des 2015; 86:723-30. [PMID: 25703929 DOI: 10.1111/cbdd.12544] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 12/15/2014] [Accepted: 01/29/2015] [Indexed: 11/28/2022]
Abstract
Hepatocellular carcinoma is the primary malignancy of the liver and is the third leading cause of cancer-related death worldwide. Despite its severity, the treatment methods for hepatocellular carcinoma are limited due to poor prognosis. This study identified the short hairpin RNA-mediated silencing of myosin VI as a potential approach for the treatment of hepatocellular carcinoma. Firstly, the expression of myosin VI was analyzed in 57 hepatocellular carcinoma samples and 10 non-neoplastic samples by immunohistochemistry. The results demonstrated that myosin VI expression was much stronger in hepatocellular carcinoma tissues than in adjacent normal tissues. Myosin VI short hairpin RNA was then transduced into HepG2 and SMMC-7721 hepatocellular carcinoma cell lines, respectively, using a lentivirus delivery system. Knockdown of myosin VI led to a significant reduction in cell proliferation and colony-forming capacity, as well as a blockade of cell cycle progression. Moreover, an obvious decrease in PRAS40 phosphorylation and a concomitant increase in p38 phosphorylation were observed in myosin VI knockdown cells, which suggest that myosin VI silencing inhibits hepatocellular carcinoma cell growth in vitro probably via inactivation of PRAS40 and activation of p38 mitogen-activated protein kinase-dependent signaling pathway. This study highlights the potential of myosin VI to be developed as a target for hepatocellular carcinoma therapy.
Collapse
Affiliation(s)
- Xiaoming Ma
- Department of General Surgery, the Second affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Jiqi Yan
- Department of Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025, China
| | - Wei Chen
- Department of General Surgery, the Second affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Peng Du
- Department of General Surgery, the Second affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Jiaming Xie
- Department of General Surgery, the Second affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Hongpei Yu
- Department of General Surgery, the Second affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Haorong Wu
- Department of General Surgery, the Second affiliated Hospital of Soochow University, Suzhou, 215004, China
| |
Collapse
|
46
|
|
47
|
Lee YH, Seo D, Choi KJ, Andersen JB, Won MA, Kitade M, Gómez-Quiroz LE, Judge AD, Marquardt JU, Raggi C, Conner EA, MacLachlan I, Factor VM, Thorgeirsson SS. Antitumor effects in hepatocarcinoma of isoform-selective inhibition of HDAC2. Cancer Res 2014; 74:4752-61. [PMID: 24958469 DOI: 10.1158/0008-5472.can-13-3531] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Histone deacetylase 2 (HDAC2) is a chromatin modifier involved in epigenetic regulation of cell cycle, apoptosis, and differentiation that is upregulated commonly in human hepatocellular carcinoma (HCC). In this study, we show that specific targeting of this HDAC isoform is sufficient to inhibit HCC progression. siRNA-mediated silencing of HDAC inhibited HCC cell growth by blocking cell-cycle progression and inducing apoptosis. These effects were associated with deregulation of HDAC-regulated genes that control cell cycle, apoptosis, and lipid metabolism, specifically, by upregulation of p27 and acetylated p53 and by downregulation of CDK6 and BCL2. We found that HDAC2 silencing in HCC cells also strongly inhibited PPARγ signaling and other regulators of glycolysis (ChREBPα and GLUT4) and lipogenesis (SREBP1C and FAS), eliciting a marked decrease in fat accumulation. Notably, systemic delivery of HDAC2 siRNA encapsulated in lipid nanoparticles was sufficient to blunt the growth of human HCC in a murine xenograft model. Our findings offer preclinical proof-of-concept for HDAC2 blockade as a systemic therapy for liver cancer.
Collapse
Affiliation(s)
- Yun-Han Lee
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland. Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, Korea.
| | - Daekwan Seo
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Kyung-Ju Choi
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, Korea
| | - Jesper B Andersen
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Min-Ah Won
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, Korea
| | - Mitsuteru Kitade
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Luis E Gómez-Quiroz
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Adam D Judge
- Tekmira Pharmaceuticals, Corp., Burnaby, British Columbia, Canada
| | - Jens U Marquardt
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Chiara Raggi
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Elizabeth A Conner
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Ian MacLachlan
- Tekmira Pharmaceuticals, Corp., Burnaby, British Columbia, Canada
| | - Valentina M Factor
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Snorri S Thorgeirsson
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
48
|
Panattoni M, Maiorino L, Lukacs A, Zentilin L, Mazza D, Sanvito F, Sitia G, Guidotti LG, Pardi R. The COP9 signalosome is a repressor of replicative stress responses and polyploidization in the regenerating liver. Hepatology 2014; 59:2331-43. [PMID: 24452456 DOI: 10.1002/hep.27028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 01/16/2014] [Indexed: 12/28/2022]
Abstract
UNLABELLED Aberrant DNA replication induced by deregulated or excessive proliferative stimuli evokes a "replicative stress response" leading to cell cycle restriction and/or apoptosis. This robust fail-safe mechanism is eventually bypassed by transformed cells, due to ill-defined epistatic interactions. The COP9 signalosome (CSN) is an evolutionarily conserved regulator of cullin ring ligases (CRLs), the largest family of ubiquitin ligases in metazoans. Conditional inactivation of the CSN in several tissues leads to activation of S- or G2-phase checkpoints resulting in irreversible cell cycle arrest and cell death. Herein we ablated COPS5, the CSNs catalytic subunit, in the liver, to investigate its role in cell cycle reentry by differentiated hepatocytes. Lack of COPS5 in regenerating livers causes substantial replicative stress, which triggers a CDKN2A-dependent genetic program leading to cell cycle arrest, polyploidy, and apoptosis. These outcomes are phenocopied by acute overexpression of c-Myc in COPS5 null hepatocytes of adult mice. CONCLUSION We propose that combined control of proto-oncogene product levels and proteins involved in DNA replication origin licensing may explain the deleterious consequences of CSN inactivation in regenerating livers and provide insight into the pathogenic role of the frequently observed overexpression of the CSN in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Martina Panattoni
- Leukocyte Biology Unit, Ospedale San Raffaele Scientific Institute, Milano, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Nanoparticle-formulated siRNA targeting integrins inhibits hepatocellular carcinoma progression in mice. Nat Commun 2014; 5:3869. [PMID: 24844798 PMCID: PMC4107318 DOI: 10.1038/ncomms4869] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 04/11/2014] [Indexed: 02/07/2023] Open
Abstract
Integrins play an important role during development, regulating cell differentiation, proliferation and survival. Here we show that knockdown of integrin subunits slows down the progression of hepatocellular carcinoma (HCC). Using nanoparticulate delivery of short interfering RNAs targeting β1 and αv integrin subunits we downregulate all integrin receptors in hepatocytes. Short-term integrin knockdown (two weeks) does not cause apparent structural or functional perturbations of normal liver tissue. Alterations in liver morphology accumulate upon sustained integrin downregulation (seven weeks). The integrin knockdown leads to significant retardation of HCC progression, reducing proliferation and increasing tumour cell death. This tumour retardation is accompanied by reduced activation of MET oncogene as well as expression of its mature form on the cell surface. Our data suggest that transformed proliferating cells from HCC are more sensitive to knockdown of integrins than normal quiescent hepatocytes, highlighting the potential of siRNA-mediated inhibition of integrins as an anti-cancer therapeutic approach.
Collapse
|
50
|
Di Martino MT, Campani V, Misso G, Gallo Cantafio ME, Gullà A, Foresta U, Guzzi PH, Castellano M, Grimaldi A, Gigantino V, Franco R, Lusa S, Cannataro M, Tagliaferri P, De Rosa G, Tassone P, Caraglia M. In vivo activity of miR-34a mimics delivered by stable nucleic acid lipid particles (SNALPs) against multiple myeloma. PLoS One 2014; 9:e90005. [PMID: 24587182 PMCID: PMC3937395 DOI: 10.1371/journal.pone.0090005] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 01/24/2014] [Indexed: 01/07/2023] Open
Abstract
Multiple myeloma (MM) is a disease with an adverse outcome and new therapeutic strategies are urgently awaited. A rising body of evidence supports the notion that microRNAs (miRNAs), master regulators of eukaryotic gene expression, may exert anti-MM activity. Here, we evaluated the activity of synthetic miR-34a in MM cells. We found that transfection of miR-34a mimics in MM cells induces a significant change of gene expression with relevant effects on multiple signal transduction pathways. We detected early inactivation of pro-survival and proliferative kinases Erk-2 and Akt followed at later time points by caspase-6 and -3 activation and apoptosis induction. To improve the in vivo delivery, we encapsulated miR-34a mimics in stable nucleic acid lipid particles (SNALPs). We found that SNALPs miR-34a were highly efficient in vitro in inhibiting growth of MM cells. Then, we investigated the activity of the SNALPs miR-34a against MM xenografts in SCID mice. We observed significant tumor growth inhibition (p<0.05) which translated in mice survival benefits (p = 0.0047). Analysis of miR-34a and NOTCH1 expression in tumor retrieved from animal demonstrated efficient delivery and gene modulation induced by SNALPs miR-34a in the absence of systemic toxicity. We here therefore provide evidence that SNALPs miR-34a may represent a promising tool for miRNA-therapeutics in MM.
Collapse
Affiliation(s)
- Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University and Medical Oncology Unit, Catanzaro, Italy
- T. Campanella Cancer Center, “Salvatore Venuta” University Campus, Catanzaro, Italy
| | - Virginia Campani
- Department of Pharmacy, Federico II University of Naples, Naples, Italy
| | - Gabriella Misso
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Maria Eugenia Gallo Cantafio
- Department of Experimental and Clinical Medicine, Magna Graecia University and Medical Oncology Unit, Catanzaro, Italy
| | - Annamaria Gullà
- Department of Experimental and Clinical Medicine, Magna Graecia University and Medical Oncology Unit, Catanzaro, Italy
| | - Umberto Foresta
- Department of Experimental and Clinical Medicine, Magna Graecia University and Medical Oncology Unit, Catanzaro, Italy
| | - Pietro Hiram Guzzi
- Department of Medical and Surgical Sciences, Laboratory of Bioinformatics Unit, “Salvatore Venuta” University Campus, Catanzaro, Italy
| | - Maria Castellano
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Anna Grimaldi
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Vincenzo Gigantino
- Pathology Unit, National Institute of Tumours of Naples “Pascale”, Naples, Italy
| | - Renato Franco
- Pathology Unit, National Institute of Tumours of Naples “Pascale”, Naples, Italy
| | - Sara Lusa
- Department of Pharmacy, Federico II University of Naples, Naples, Italy
| | - Mario Cannataro
- Department of Medical and Surgical Sciences, Laboratory of Bioinformatics Unit, “Salvatore Venuta” University Campus, Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University and Medical Oncology Unit, Catanzaro, Italy
- T. Campanella Cancer Center, “Salvatore Venuta” University Campus, Catanzaro, Italy
| | - Giuseppe De Rosa
- Department of Pharmacy, Federico II University of Naples, Naples, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University and Medical Oncology Unit, Catanzaro, Italy
- T. Campanella Cancer Center, “Salvatore Venuta” University Campus, Catanzaro, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|