1
|
Jia GG, Lu LX, Li B, Li CY, Zheng Y, Zhang JC, He YJ, Xu-Shi, Yu XH. lncRNA-NEF regulates hepatic stellate cells proliferation, cell cycle, apoptosis and ECM synthesis through the ERK1/2/c-Fos axis. Exp Cell Res 2025; 444:114361. [PMID: 39622465 DOI: 10.1016/j.yexcr.2024.114361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/01/2024] [Accepted: 11/27/2024] [Indexed: 01/14/2025]
Abstract
In this study, we investigated the role of lncRNA-NEF in modulating hepatic stellate cell (HSC) activation, a key process in liver fibrosis. Using the GSE78160 dataset, we identified lncRNA-NEF as downregulated in liver cirrhosis patients. Gene Ontology and KEGG analyses implicated it in transcriptional regulation and cell cycle control. We established an activated HSC model with TGF-β1-treated LX-2 cells and employed RT-qPCR and Western blot to assess lncRNA-NEF and ERK1/2 expression. Lentiviral transfection was used to overexpress lncRNA-NEF in activated LX-2 cells, and its effects on proliferation, apoptosis, and cell cycle were evaluated using EdU staining, CCK-8, Annexin-V PE/7-AAD, TUNEL, and PI-FACS analysis. Overexpression of lncRNA-NEF led to reduced cell proliferation, increased apoptosis, and cell cycle arrest at the S and G2/M phases. We also observed a decrease in ERK1/2, c-Fos, Collagen I, α-SMA, and Bcl-2 expression, and an increase in Caspase-3 expression, as confirmed by Western blot. These results suggest that lncRNA-NEF regulates HSC activation via the ERK1/2/c-Fos axis, potentially offering a therapeutic target for antifibrotic drug development. Our findings provide a molecular basis for understanding the role of lncRNAs in liver fibrosis and highlight the potential of lncRNA-NEF as a novel antifibrotic target.
Collapse
Affiliation(s)
- Gang-Gang Jia
- Gansu University of Chinese Medicine, Lanzhou, China; Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of PLA, Lanzhou, China.
| | - Li-Xia Lu
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of PLA, Lanzhou, China.
| | - Bin- Li
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of PLA, Lanzhou, China.
| | - Chu-Yi Li
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of PLA, Lanzhou, China.
| | - Ying- Zheng
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of PLA, Lanzhou, China.
| | - Jiu-Cong Zhang
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of PLA, Lanzhou, China.
| | - Yu-Jing He
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of PLA, Lanzhou, China.
| | - Xu-Shi
- Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of PLA, Lanzhou, China.
| | - Xiao-Hui Yu
- Gansu University of Chinese Medicine, Lanzhou, China; Department of Gastroenterology, The 940th Hospital of Joint Service Logistics Support Force of PLA, Lanzhou, China.
| |
Collapse
|
2
|
Zhang H, Li Q, Guo X, Wu H, Hu C, Liu G, Yu T, Hu X, Qiu Q, Guo G, She J, Chen Y. MGMT activated by Wnt pathway promotes cisplatin tolerance through inducing slow-cycling cells and nonhomologous end joining in colorectal cancer. J Pharm Anal 2024; 14:100950. [PMID: 39027911 PMCID: PMC11255892 DOI: 10.1016/j.jpha.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 07/20/2024] Open
Abstract
Chemotherapy resistance plays a pivotal role in the prognosis and therapeutic failure of patients with colorectal cancer (CRC). Cisplatin (DDP)-resistant cells exhibit an inherent ability to evade the toxic chemotherapeutic drug effects which are characterized by the activation of slow-cycle programs and DNA repair. Among the elements that lead to DDP resistance, O 6-methylguanine (O 6-MG)-DNA-methyltransferase (MGMT), a DNA-repair enzyme, performs a quintessential role. In this study, we clarify the significant involvement of MGMT in conferring DDP resistance in CRC, elucidating the underlying mechanism of the regulatory actions of MGMT. A notable upregulation of MGMT in DDP-resistant cancer cells was found in our study, and MGMT repression amplifies the sensitivity of these cells to DDP treatment in vitro and in vivo. Conversely, in cancer cells, MGMT overexpression abolishes their sensitivity to DDP treatment. Mechanistically, the interaction between MGMT and cyclin dependent kinase 1 (CDK1) inducing slow-cycling cells is attainted via the promotion of ubiquitination degradation of CDK1. Meanwhile, to achieve nonhomologous end joining, MGMT interacts with XRCC6 to resist chemotherapy drugs. Our transcriptome data from samples of 88 patients with CRC suggest that MGMT expression is co-related with the Wnt signaling pathway activation, and several Wnt inhibitors can repress drug-resistant cells. In summary, our results point out that MGMT is a potential therapeutic target and predictive marker of chemoresistance in CRC.
Collapse
Affiliation(s)
- Haowei Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Qixin Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiaolong Guo
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Hong Wu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Chenhao Hu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Gaixia Liu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Tianyu Yu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiake Hu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Quanpeng Qiu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Gang Guo
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Junjun She
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yinnan Chen
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
3
|
Zhu Y, Dai Z. HSP90: A promising target for NSCLC treatments. Eur J Pharmacol 2024; 967:176387. [PMID: 38311278 DOI: 10.1016/j.ejphar.2024.176387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/15/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
The emergence of targeted therapies and immunotherapies has improved the overall survival of patients with nonsmall cell lung cancer (NSCLC), but the 5-year survival rate remains low. New drugs are needed to overcome this dilemma. Moreover, the significant correlation between various client proteins of heat-shock protein (HSP) 90 and tumor occurrence, progression, and drug resistance suggests that HSP90 is a potential therapeutic target for NSCLC. However, the outcomes of clinical trials for HSP90 inhibitors have been disappointing, indicating significant toxicity of these drugs and that further screening of the beneficiary population is required. NSCLC patients with oncogenic-driven gene mutations or those at advanced stages who are resistant to multi-line treatments may benefit from HSP90 inhibitors. Enhancing the therapeutic efficacy and reducing the toxicity of HSP90 inhibitors can be achieved via the optimization of their drug structure, using them in combination therapies with low-dose HSP90 inhibitors and other drugs, and via targeted administration to tumor lesions. Here, we provide a review of the recent research on the role of HSP90 in NSCLC and summarize relevant studies of HSP90 inhibitors in NSCLC.
Collapse
Affiliation(s)
- Yue Zhu
- Department of Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116021, Liaoning Province, China
| | - Zhaoxia Dai
- Department of Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116021, Liaoning Province, China.
| |
Collapse
|
4
|
Luo H, Wu X, Huo B, Liu L, Jiang DS, Yi X. The roles of METTL3 on autophagy and proliferation of vascular smooth muscle cells are mediated by mTOR rather than by CDK1. Cell Div 2023; 18:13. [PMID: 37559091 PMCID: PMC10411010 DOI: 10.1186/s13008-023-00096-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/02/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Aberrant proliferation of vascular smooth muscle cells (VSMCs) is the cause of neointima formation followed by vascular injury. Autophagy is involved in this pathological process, but its function is controversial. Recently, we found that methyltransferase like 3 (METTL3) inhibited VSMC proliferation by activating autophagosome formation. Moreover, we also demonstrated that METTL3 reduced the levels of phosphorylated mammalian target of rapamycin (p-mTOR) and cyclin dependent kinase 1 (p-CDK1/CDC2), which were critical for autophagy and proliferation regulation. However, whether mTOR and CDK1 mediated the function of METTL3 on autophagy and proliferation in VSMCs remains unknown. RESULTS We showed that the activator of mTOR, MHY1485 largely reversed the effects of METTL3 overexpression on VSMC autophagy and proliferation. Rapamycin, the inhibitor of mTOR, obviously nullified the pro-proliferation effects of METTL3 knockdown by activating autophagy in VSMCs. Unexpectedly, mTOR did not contribute to the impacts of METTL3 on migration and phenotypic switching of VSMCs. On the other hand, by knockdown of CDK1 in VSMC with METTL3 deficiency, we demonstrated that CDK1 was involved in METTL3-regulated proliferation of VSMCs, but this effect was not mediated by autophagy. CONCLUSIONS We concluded that mTOR but not CDK1 mediated the role of METTL3 on VSMC proliferation and autophagy.
Collapse
Affiliation(s)
- Hanshen Luo
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, Hubei, China
| | - Xingliang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, China
| | - Bo Huo
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, Hubei, China
| | - Liyuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, China
| | - Ding-Sheng Jiang
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| | - Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, China.
| |
Collapse
|
5
|
Peng X, Khan Z, Liu XM, Deng SL, Fang YG, Zhang M, Su XH, Xing LX, Yan XR. Embryonic Development of Parthenogenetic and Sexual Eggs in Lower Termites. INSECTS 2023; 14:640. [PMID: 37504646 PMCID: PMC10380263 DOI: 10.3390/insects14070640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/29/2023]
Abstract
Worldwide, termites are one of few social insects. In this research, the stages of embryonic development in the parthenogenetic and sexual eggs of Reticulitermes aculabialis and R. flaviceps were observed and described. In R. flaviceps, the egg development of the FF and FM groups happened during the early phases of development, whereas in R. aculabialis, this appeared mainly during the late phase of development. The variance in the number of micropyles between the R. flaviceps FF colony type and the R. aculabialis FF colony type was statistically significant. Five stages of egg development were found in both types of R. aculabialis but only the sexual eggs of R. flaviceps. In R. flaviceps, 86% of the parthenogenetic eggs stopped growing during the blastoderm development, with the yolk cell assembling frequently in the center of the egg. According to the results of the single-cell transcriptome sequencing, we investigated the egg-to-larval expression level of genes (pka, map2k1, mapk1/3, hgk, mkp, and pax6) and indicated that the levels of essential gene expression in RaFF were considerably higher than in RfFF (p < 0.05). We also discovered that the oocyte cleavage rate in the FF colony type was considerably lower in R. flaviceps compared to R. aculabialis, which gave rise to a smaller number of mature oocytes in R. flaviceps. During ovulation in both species, oocytes underwent activation and one or two cleavage events, but the development of unfertilized eggs ceased in R. flaviceps. It was shown that termite oocyte and embryonic development were heavily influenced by genes with significant expressions. Results from the databases KEGG, COG, and GO unigenes revealed the control of numerous biological processes. This study is the first to complete a database of parthenogenetic and sexual eggs of R. flaviceps and R. aculabialis.
Collapse
Affiliation(s)
- Xin Peng
- College of Life Sciences, Northwest University, Xi’an 710069, China; (X.P.); (Z.K.); (X.-M.L.); (S.-L.D.); (Y.-G.F.); (M.Z.); (X.-H.S.)
| | - Zahid Khan
- College of Life Sciences, Northwest University, Xi’an 710069, China; (X.P.); (Z.K.); (X.-M.L.); (S.-L.D.); (Y.-G.F.); (M.Z.); (X.-H.S.)
- Zoology Department, University of Swabi, Swabi 23561, Khyber Pakhtunkhwa, Pakistan
| | - Xiao-Min Liu
- College of Life Sciences, Northwest University, Xi’an 710069, China; (X.P.); (Z.K.); (X.-M.L.); (S.-L.D.); (Y.-G.F.); (M.Z.); (X.-H.S.)
| | - Shi-Lin Deng
- College of Life Sciences, Northwest University, Xi’an 710069, China; (X.P.); (Z.K.); (X.-M.L.); (S.-L.D.); (Y.-G.F.); (M.Z.); (X.-H.S.)
| | - Yong-Gang Fang
- College of Life Sciences, Northwest University, Xi’an 710069, China; (X.P.); (Z.K.); (X.-M.L.); (S.-L.D.); (Y.-G.F.); (M.Z.); (X.-H.S.)
| | - Min Zhang
- College of Life Sciences, Northwest University, Xi’an 710069, China; (X.P.); (Z.K.); (X.-M.L.); (S.-L.D.); (Y.-G.F.); (M.Z.); (X.-H.S.)
| | - Xiao-Hong Su
- College of Life Sciences, Northwest University, Xi’an 710069, China; (X.P.); (Z.K.); (X.-M.L.); (S.-L.D.); (Y.-G.F.); (M.Z.); (X.-H.S.)
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an 710069, China
- Xi’an Brand of Chinese Academy of Sciences, Xi’an 710043, China
| | - Lian-Xi Xing
- College of Life Sciences, Northwest University, Xi’an 710069, China; (X.P.); (Z.K.); (X.-M.L.); (S.-L.D.); (Y.-G.F.); (M.Z.); (X.-H.S.)
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an 710069, China
- Xi’an Brand of Chinese Academy of Sciences, Xi’an 710043, China
| | - Xing-Rong Yan
- College of Life Sciences, Northwest University, Xi’an 710069, China; (X.P.); (Z.K.); (X.-M.L.); (S.-L.D.); (Y.-G.F.); (M.Z.); (X.-H.S.)
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an 710069, China
- Xi’an Brand of Chinese Academy of Sciences, Xi’an 710043, China
| |
Collapse
|
6
|
Wei S, Xing J, Lu K, Wang K, Yu W. NPM3 as a novel oncogenic factor and poor prognostic marker contributes to cell proliferation and migration in lung adenocarcinoma. Hereditas 2023; 160:27. [PMID: 37254219 PMCID: PMC10230701 DOI: 10.1186/s41065-023-00289-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/21/2023] [Indexed: 06/01/2023] Open
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-related deaths worldwide, and despite recent advances in targeted therapies and immunotherapies, the clinical benefit remains limited. Therefore, there is an urgent need to further investigate the molecular mechanisms underlying lung cancer. The aim of this study was to investigate the expression and function of NPM3 in the tumor microenvironment of lung adenocarcinoma (LUAD). METHODS We utilized bioinformatics tools and databases, including UALCAN, GEPIA2, HPA, and Sangerbox, to analyze NPM3 expression in LUAD samples and its association with prognosis and mutational landscape. NPM3 expression in various cell types was assessed at the single cell level using the TISCH database. We also used algorithms such as TIMER and EPIC to explore the crosstalk between NPM3 expression and immune features. KEGG enrichment analysis was performed to identify potential signaling pathways of NPM3. Finally, we employed siRNA knockdown strategy to investigate the effect of NPM3 on LUAD cell proliferation and migration in vitro. RESULTS NPM3 was significantly upregulated in LUAD tissues and was strongly associated with poor prognosis and TP53 gene mutations. Single-cell sequencing analysis revealed that NPM3 was expressed in immune cells (dendritic cells and monocytes/macrophages) in the tumor microenvironment. Moreover, NPM3 expression was negatively associated with immune B cell and CD4 T cell infiltration, as well as with several immune-related genes (including CCL22, CXCR2, CX3CR1, CCR6, HLA-DOA, HLA-DQA2). KEGG enrichment analysis indicated that NPM3 expression was associated with cell cycle, CAMs, and NSCLC pathway genes. Finally, in vitro experiments showed that NPM3 knockdown inhibited LUAD cell proliferation and migration in NCI-H1299 and SPC-A1 cells, and suppressed the expression of CCNA2 and MAD2L1. CONCLUSION Elevated NPM3 expression predicts poor clinical outcome and an immunosuppressive microenvironment in LUAD tissues. NPM3 promotes LUAD progression by promoting cell proliferation and migration, and targeting NPM3 may represent a novel therapeutic strategy for LUAD.
Collapse
Affiliation(s)
- Shan Wei
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, 322000, Zhejiang, People's Republic of China
- Department of Respiratory and Critical Care Medicine, The Affiliated People's Hospital of Ningbo University (Ningbo Yinzhou People's Hospital), No.251, Baizhang Road, Ningbo, 315040, Zhejiang, People's Republic of China
| | - Jing Xing
- Department of Respiratory and Critical Care Medicine, The Affiliated People's Hospital of Ningbo University (Ningbo Yinzhou People's Hospital), No.251, Baizhang Road, Ningbo, 315040, Zhejiang, People's Republic of China
- Ningbo University School of Medicine, Zhejiang Province, Ningbo, People's Republic of China
| | - Kaining Lu
- Department of Urology, The Affiliated First Hospital of Ningbo University (Ningbo First Hospital), No.59, Liuting Street, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, 322000, Zhejiang, People's Republic of China
| | - Wanjun Yu
- Department of Respiratory and Critical Care Medicine, The Affiliated People's Hospital of Ningbo University (Ningbo Yinzhou People's Hospital), No.251, Baizhang Road, Ningbo, 315040, Zhejiang, People's Republic of China.
| |
Collapse
|
7
|
Chowdhury SR, Koley T, Singh M, Samath EA, Kaur P. Association of Hsp90 with p53 and Fizzy related homolog (Fzr) synchronizing Anaphase Promoting Complex (APC/C): An unexplored ally towards oncogenic pathway. Biochim Biophys Acta Rev Cancer 2023; 1878:188883. [PMID: 36972769 DOI: 10.1016/j.bbcan.2023.188883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/29/2023]
Abstract
The intricate molecular interactions leading to the oncogenic pathway are the consequence of cell cycle modification controlled by a bunch of cell cycle regulatory proteins. The tumor suppressor and cell cycle regulatory proteins work in coordination to maintain a healthy cellular environment. The integrity of this cellular protein pool is perpetuated by heat shock proteins/chaperones, which assist in proper protein folding during normal and cellular stress conditions. Among these versatile groups of chaperone proteins, Hsp90 is one of the significant ATP-dependent chaperones that aid in stabilizing many tumor suppressors and cell cycle regulator protein targets. Recently, studies have revealed that in cancerous cell lines, Hsp90 stabilizes mutant p53, 'the guardian of the genome.' Hsp90 also has a significant impact on Fzr, an essential regulator of the cell cycle having an important role in the developmental process of various organisms, including Drosophila, yeast, Caenorhabditis elegans, and plants. During cell cycle progression, p53 and Fzr coordinately regulate the Anaphase Promoting Complex (APC/C) from metaphase to anaphase transition up to cell cycle exit. APC/C mediates proper centrosome function in the dividing cell. The centrosome acts as the microtubule organizing center for the correct segregation of the sister chromatids to ensure perfect cell division. This review examines the structure of Hsp90 and its co-chaperones, which work in synergy to stabilize proteins such as p53 and Fizzy-related homolog (Fzr) to synchronize the Anaphase Promoting Complex (APC/C). Dysfunction of this process activates the oncogenic pathway leading to the development of cancer. Additionally, an overview of current drugs targeting Hsp90 at various phases of clinical trials has been included.
Collapse
Affiliation(s)
- Sanghati Roy Chowdhury
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Tirthankar Koley
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Mandeep Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | | | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
8
|
Zhang Z, Wang X, Nie P, Qin Y, Shi J, Xu S. DEPDC1B promotes development of cholangiocarcinoma through enhancing the stability of CDK1 and regulating malignant phenotypes. Front Oncol 2022; 12:842205. [PMID: 36568241 PMCID: PMC9769124 DOI: 10.3389/fonc.2022.842205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 11/04/2022] [Indexed: 12/12/2022] Open
Abstract
Cholangiocarcinoma (CCA) is the second most common primary tumor of the hepatobiliary system. At present, the therapeutic efficiency of cholangiocarcinoma is fairly low and the prognosis is poor. The root cause is that the molecular mechanism of the occurrence and development of CCA is largely unclear. This work intended to clarify the role of DEP domain-containing protein 1B (DEPDC1B) in the progress of CCA through cellular biology research strategies and further clarify the molecular mechanism of CCA. Clinical tissue-related detection showed that the expression level of DEPDC1B in tumor tissues was significantly higher than that in normal tissues and was positively correlated with tumor grade. Knockdown of the endogenous DEPDC1B of CCA cells can significantly inhibit cell proliferation and migration, while promoting cell apoptosis and blocking the cell cycle. DEPDC1B overexpression induced the opposite effects. Studies in animal models also showed that the downregulation of DEPDC1B can reduce the tumorigenicity of CCA cells. In addition, through gene profiling analysis and molecular biology studies, we found that CDK1 may be an important downstream mediator of DEPDC1B, the protein stability of which was significantly decreased through the ubiquitin-proteasome system in DEPDC1B knockdown cells. Moreover, knockdown of CDK1 can weaken the promotion of CCA caused by DEPDC1B overexpression. In summary, our research showed that DEPDC1B plays an important role in the development of CCA and its targeted inhibition may become one of the important methods to inhibit the progress of CCA.
Collapse
Affiliation(s)
- Zhenhai Zhang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xinxing Wang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Peihua Nie
- Department of Ophthalmology and Otorhinolaryngology, Shandong Provincial Third hospital, Jinan, Shandong, China
| | - Yejun Qin
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Junping Shi
- Medical Department, OrigiMed, Shanghai, China
| | - Shifeng Xu
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
9
|
Chen JH, Wu CH, Jheng JR, Chao CT, Huang JW, Hung KY, Liu SH, Chiang CK. The down-regulation of XBP1, an unfolded protein response effector, promotes acute kidney injury to chronic kidney disease transition. J Biomed Sci 2022; 29:46. [PMID: 35765067 PMCID: PMC9241279 DOI: 10.1186/s12929-022-00828-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/16/2022] [Indexed: 11/10/2022] Open
Abstract
Background The activation of the unfolded protein response (UPR) is closely linked to the pathogenesis of renal injuries. However, the role of XBP1, a crucial regulator of adaptive UPR, remains unclear during the transition from acute kidney injury (AKI) to chronic kidney disease (CKD). Methods We characterized XBP1 expressions in different mouse models of kidney injuries, including unilateral ischemia–reperfusion injury (UIRI), unilateral ureteral obstruction, and adenine-induced CKD, followed by generating proximal tubular XBP1 conditional knockout (XBP1cKO) mice for examining the influences of XBP1. Human proximal tubular epithelial cells (HK-2) were silenced of XBP1 to conduct proteomic analysis and investigate the underlying mechanism. Results We showed a tripartite activation of UPR in injured kidneys. XBP1 expressions were attenuated after AKI and inversely correlated with the severity of post-AKI renal fibrosis. XBP1cKO mice exhibited more severe renal fibrosis in the UIRI model than wide-type littermates. Silencing XBP1 induced HK-2 cell cycle arrest in G2M phase, inhibited cell proliferation, and promoted TGF-β1 secretion. Proteomic analysis identified TNF receptor associated protein 1 (Trap1) as the potential downstream target transcriptionally regulated by XBP1s. Trap1 overexpression can alleviate silencing XBP1 induced profibrotic factor expressions and cell cycle arrest. Conclusion The loss of XBP1 in kidney injury was profibrotic, and the process was mediated by autocrine and paracrine regulations in combination. The present study identified the XBP1-Trap1 axis as an instrumental mechanism responsible for post-AKI fibrosis, which is a novel regulatory pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s12929-022-00828-9.
Collapse
Affiliation(s)
- Jia-Huang Chen
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, No.1 Jen Ai road section 1, Taipei, 100, Taiwan
| | - Chia-Hsien Wu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, No.1 Jen Ai road section 1, Taipei, 100, Taiwan
| | - Jia-Rong Jheng
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, No.1 Jen Ai road section 1, Taipei, 100, Taiwan.,Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Ter Chao
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, No.1 Jen Ai road section 1, Taipei, 100, Taiwan.,Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jenq-Wen Huang
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuan-Yu Hung
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shing-Hwa Liu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, No.1 Jen Ai road section 1, Taipei, 100, Taiwan
| | - Chih-Kang Chiang
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, No.1 Jen Ai road section 1, Taipei, 100, Taiwan. .,Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei, Taiwan. .,Center for Biotechnology, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
10
|
Wengert LA, Backe SJ, Bourboulia D, Mollapour M, Woodford MR. TRAP1 Chaperones the Metabolic Switch in Cancer. Biomolecules 2022; 12:biom12060786. [PMID: 35740911 PMCID: PMC9221471 DOI: 10.3390/biom12060786] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/02/2022] [Accepted: 06/02/2022] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial function is dependent on molecular chaperones, primarily due to their necessity in the formation of respiratory complexes and clearance of misfolded proteins. Heat shock proteins (Hsps) are a subset of molecular chaperones that function in all subcellular compartments, both constitutively and in response to stress. The Hsp90 chaperone TNF-receptor-associated protein-1 (TRAP1) is primarily localized to the mitochondria and controls both cellular metabolic reprogramming and mitochondrial apoptosis. TRAP1 upregulation facilitates the growth and progression of many cancers by promoting glycolytic metabolism and antagonizing the mitochondrial permeability transition that precedes multiple cell death pathways. TRAP1 attenuation induces apoptosis in cellular models of cancer, identifying TRAP1 as a potential therapeutic target in cancer. Similar to cytosolic Hsp90 proteins, TRAP1 is also subject to post-translational modifications (PTM) that regulate its function and mediate its impact on downstream effectors, or ‘clients’. However, few effectors have been identified to date. Here, we will discuss the consequence of TRAP1 deregulation in cancer and the impact of post-translational modification on the known functions of TRAP1.
Collapse
Affiliation(s)
- Laura A. Wengert
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (L.A.W.); (S.J.B.); (D.B.); (M.M.)
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Sarah J. Backe
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (L.A.W.); (S.J.B.); (D.B.); (M.M.)
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Dimitra Bourboulia
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (L.A.W.); (S.J.B.); (D.B.); (M.M.)
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Mehdi Mollapour
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (L.A.W.); (S.J.B.); (D.B.); (M.M.)
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Mark R. Woodford
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (L.A.W.); (S.J.B.); (D.B.); (M.M.)
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Correspondence:
| |
Collapse
|
11
|
Yang L, Xiong H, Li X, Li Y, Zhou H, Lin X, Chan TF, Li R, Lai KP, Chen X. Network Pharmacology and Comparative Transcriptome Reveals Biotargets and Mechanisms of Curcumol Treating Lung Adenocarcinoma Patients With COVID-19. Front Nutr 2022; 9:870370. [PMID: 35520289 PMCID: PMC9063984 DOI: 10.3389/fnut.2022.870370] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/16/2022] [Indexed: 12/31/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has led to 4,255,892 deaths worldwide. Although COVID-19 vaccines are available, mutant forms of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have reduced the effectiveness of vaccines. Patients with cancer are more vulnerable to COVID-19 than patients without cancer. Identification of new drugs to treat COVID-19 could reduce mortality rate, and traditional Chinese Medicine(TCM) has shown potential in COVID-19 treatment. In this study, we focused on lung adenocarcinoma (LUAD) patients with COVID-19. We aimed to investigate the use of curcumol, a TCM, to treat LUAD patients with COVID-19, using network pharmacology and systematic bioinformatics analysis. The results showed that LUAD and patients with COVID-19 share a cluster of common deregulated targets. The network pharmacology analysis identified seven core targets (namely, AURKA, CDK1, CCNB1, CCNB2, CCNE1, CCNE2, and TTK) of curcumol in patients with COVID-19 and LUAD. Clinicopathological analysis of these targets demonstrated that the expression of these targets is associated with poor patient survival rates. The bioinformatics analysis further highlighted the involvement of this target cluster in DNA damage response, chromosome stability, and pathogenesis of LUAD. More importantly, these targets influence cell-signaling associated with the Warburg effect, which supports SARS-CoV-2 replication and inflammatory response. Comparative transcriptomic analysis on in vitro LUAD cell further validated the effect of curcumol for treating LUAD through the control of cell cycle and DNA damage response. This study supports the earlier findings that curcumol is a potential treatment for patients with LUAD and COVID-19.
Collapse
Affiliation(s)
- Lu Yang
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
| | - Hao Xiong
- Guilin Center for Disease Control and Prevention, Guilin, China
| | - Xin Li
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Yu Li
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
| | - Huanhuan Zhou
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
| | - Xiao Lin
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ting Fung Chan
- State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Rong Li
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
- *Correspondence: Rong Li
| | - Keng Po Lai
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, China
- Keng Po Lai
| | - Xu Chen
- Department of Pharmacy, Guilin Medical University, Guilin, China
- Xu Chen ;
| |
Collapse
|
12
|
TRAP1 inhibits MIC60 ubiquitination to mitigate the injury of cardiomyocytes and protect mitochondria in extracellular acidosis. Cell Death Dis 2021; 7:389. [PMID: 34907169 PMCID: PMC8671480 DOI: 10.1038/s41420-021-00786-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/23/2021] [Accepted: 12/02/2021] [Indexed: 12/19/2022]
Abstract
Extracellular acidosis-induced mitochondrial damage of cardiomyocytes leads to cardiac dysfunction, but no detailed mechanism or efficient therapeutic target has been reported. Here we found that the protein levels of MIC60 were decreased in H9C2 cells and heart tissues in extracellular acidosis, which caused mitochondrial damage and cardiac dysfunction. Overexpression of MIC60 maintains H9C2 cells viability, increases ATP production and mitochondrial membrane potential, mitigates the disruptions of mitochondrial structure and cardiac injury. Mechanistically, extracellular acidosis excessively promoted MIC60 ubiquitin-dependent degradation. TRAP1 mitigated acidosis-induced mitochondrial impairments and cardiac injury by directly interacting with MIC60 to decrease its ubiquitin-dependent degradation in extracellular acidosis.
Collapse
|
13
|
Ma J, Chen X, Zhu X, Pan Z, Hao W, Li D, Zheng Q, Tang X. Luteolin potentiates low-dose oxaliplatin-induced inhibitory effects on cell proliferation in gastric cancer by inducing G 2/M cell cycle arrest and apoptosis. Oncol Lett 2021; 23:16. [PMID: 34820015 PMCID: PMC8607327 DOI: 10.3892/ol.2021.13134] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/13/2021] [Indexed: 01/02/2023] Open
Abstract
Although the reduction of oxaliplatin doses may alleviate deleterious side effects of gastrointestinal and gynecological cancer treatment, it also limits the anticancer therapeutic effects. As a high-efficient and low-priced herbal medicine ingredient, luteolin is an agent with a broad spectrum of anticancer activities and acts as a potential enhancer of therapeutic effects of chemotherapy agents in cancer treatment. This study focused on the antitumor effects and mechanism of combined treatment with luteolin and oxaliplatin on a mouse forestomach carcinoma (MFC) cell line. The study used CCK-8 assay, flow cytometry, Annexin V-FITC/PI double staining assay, reactive oxygen species testing assay, mitochondrial membrane potential testing assay, and western blot assay. The results showed that luteolin and oxaliplatin exerted synergistic effects on inhibiting MFC cell proliferation by inducing G2/M cell cycle arrest and apoptosis. Inhibiting the tumor necrosis factor receptor-associated protein 1/phosphorylated-extracellular-regulated protein kinases1/2/cell division cycle 25 homolog C/cyclin-dependent kinase-1/cyclin B1 pathway was indispensable to the combined treatment with luteolin and oxaliplatin to induce G2/M cell cycle arrest. In addition, luteolin increased oxidative stress in MFC cells treated with a low dose of oxaliplatin. The combined therapy damaged mitochondrial membrane potential and regulated BCL-2-associated X protein and B-cell lymphoma 2 protein expression, leading to apoptosis. Findings of the present study suggest that luteolin may be a qualified chemotherapy enhancer to potentiate the anticancer effects of low-dose oxaliplatin in MFC cells. This work provides a theoretical foundation for future research on applications of luteolin in clinical chemotherapy.
Collapse
Affiliation(s)
- Jun Ma
- College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong 266003, P.R. China.,School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Xiaojie Chen
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Xuejie Zhu
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Zhaohai Pan
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Wenjin Hao
- School of Life Sciences, Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Defang Li
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Qiusheng Zheng
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China.,School of Pharmacy, Shihezi University, Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Ministry of Education, School of Pharmacy, Shihezi, Xinjiang 832002, P.R. China
| | - Xuexi Tang
- College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
14
|
Kłos P, Dabravolski SA. The Role of Mitochondria Dysfunction in Inflammatory Bowel Diseases and Colorectal Cancer. Int J Mol Sci 2021; 22:11673. [PMID: 34769108 PMCID: PMC8584106 DOI: 10.3390/ijms222111673] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 12/30/2022] Open
Abstract
Inflammatory bowel disease (IBD) is one of the leading gut chronic inflammation disorders, especially prevalent in Western countries. Recent research suggests that mitochondria play a crucial role in IBD development and progression to the more severe disease-colorectal cancer (CRC). In this review, we focus on the role of mitochondrial mutations and dysfunctions in IBD and CRC. In addition, main mitochondria-related molecular pathways involved in IBD to CRC transition are discussed. Additionally, recent publications dedicated to mitochondria-targeted therapeutic approaches to cure IBD and prevent CRC progression are discussed.
Collapse
Affiliation(s)
- Patrycja Kłos
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 72 Al. Powstańców Wlkp., 70-111 Szczecin, Poland;
| | - Siarhei A. Dabravolski
- Department of Clinical Diagnostics, Vitebsk State Academy of Veterinary Medicine [UO VGAVM], 7/11 Dovatora Str., 210026 Vitebsk, Belarus
| |
Collapse
|
15
|
Chen ZX, Li GS, Yang LH, Liu HC, Qin GM, Shen L, He WY, Gan TQ, Li JJ. Upregulation of BIRC5 plays essential role in esophageal squamous cell carcinoma. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:6941-6960. [PMID: 34517565 DOI: 10.3934/mbe.2021345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the most common cancers in the world, the detection and prognosis of which are still unsatisfactory. Thus, it is essential to explore the factors that may identify ESCC and evaluate the prognosis of ESCC patients. RESULTS Both protein and mRNA expression levels of BIRC5 are upregulated in ESCC group rather than non-ESCC group (standardized mean difference > 0). BIRC5 mRNA expression is related to the age, tumor location, lymph node stage and clinical stage of ESCC patients (p < 0.05). BIRC5 expression makes it feasible to distinguish ESCC from non-ESCC (area under the curve > 0.9), and its high expression is related to poor prognosis of ESCC patients (restrictive survival time difference = -0.036, p < 0.05). BIRC5 may play an important role in ESCC by influencing the cell cycle pathway, and CDK1, MAD2L and CDC20 may be the hub genes of this pathway. The transcription factors-MAZ and TFPD1 -are likely to regulate the transcription of BIRC5, which may be one of the factors for the high expression of BIRC5 in ESCC. CONCLUSIONS The current study shows that upregulation of BIRC5 may have essential clinical value in ESCC, and contributes to the understanding of the pathogenesis of ESCC.
Collapse
Affiliation(s)
- Zu-Xuan Chen
- Department of Medical Oncology, Second Affiliated Hospital of Guangxi Medical University, 166 DaxueXi Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, P. R. China
| | - Guo-Sheng Li
- Department of Medical Oncology, Second Affiliated Hospital of Guangxi Medical University, 166 DaxueXi Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, P. R. China
| | - Li-Hua Yang
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, P. R. China
| | - He-Chuan Liu
- Department of Medical Oncology, Second Affiliated Hospital of Guangxi Medical University, 166 DaxueXi Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, P. R. China
| | - Guang-Mei Qin
- Department of Medical Oncology, Second Affiliated Hospital of Guangxi Medical University, 166 DaxueXi Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, P. R. China
| | - Lang Shen
- Department of Medical Oncology, Second Affiliated Hospital of Guangxi Medical University, 166 DaxueXi Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, P. R. China
| | - Wei-Ying He
- Department of Medical Oncology, Second Affiliated Hospital of Guangxi Medical University, 166 DaxueXi Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, P. R. China
| | - Ting-Qing Gan
- Department of Medical Oncology, Second Affiliated Hospital of Guangxi Medical University, 166 DaxueXi Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, P. R. China
| | - Jian-Jun Li
- Department of General Surgery, Second Affiliated Hospital of Guangxi Medical University, 166 DaxueXi Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, P. R. China
| |
Collapse
|
16
|
Yang X, Tian M, Zhang W, Chai T, Shen Z, Kang M, Lin J. Identification of potential core genes in esophageal carcinoma using bioinformatics analysis. Medicine (Baltimore) 2021; 100:e26428. [PMID: 34232175 PMCID: PMC8270608 DOI: 10.1097/md.0000000000026428] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 06/03/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a common human malignancy worldwide. The tumorigenesis mechanism in ESCC is unclear. MATERIALS AND METHODS To explore potential therapeutic targets for ESCC, we analyzed 3 microarray datasets (GSE20347, GSE38129, and GSE67269) derived from the gene expression omnibus (GEO) database. Then, the GEO2R tool was used to screen out differently expressed genes (DEGs) between ESCC and normal tissue. Gene ontology function and kyoto encyclopedia of genes and genomes pathway enrichment analysis were performed using the database for annotation, visualization and integrated discovery to identify the pathways and functional annotation of DEGs. Protein-protein interaction of these DEGs was analyzed based on the search tool for the retrieval of interacting genes database and visualized by Cytoscape software. In addition, we used encyclopedia of RNA interactomes (ENCORI), gene expression profiling interactive analysis (GEPIA), and the human protein atlas to confirm the expression of hub genes in ESCC. Finally, GEPIA was used to evaluate the prognostic value of hub genes expression in ESCC patients and we estimated the associations between hub genes expression and immune cell populations (B Cell, CD8+ T Cell, CD4+ T Cell, Macrophage, Neutrophil, and Dendritic Cell) in esophageal carcinoma (ESCA) using tumor immune estimation resource (TIMER). RESULTS In this study, 707 DEGs (including 385 upregulated genes and 322 downregulated genes) and 6 hub genes (cyclin B1 [CCNB1], cyclin dependent kinase 1 [CDK1], aurora kinase A [AURKA], ubiquitin conjugating enzyme E2C [UBE2C], cyclin A2 [CCNA2], and cell division cycle 20 [CDC20]) were identified. All of the 6 hub genes were highly expressed in ESCC tissues. Among of them, only CCNB1 and CDC20 were associated with stage of ESCC and all of them were not associated with survival time of patients. CONCLUSION DEGs and hub genes were confirmed in our study, providing a thorough, scientific and comprehensive research goals for the pathogenesis of ESCC.
Collapse
Affiliation(s)
| | - Mengyue Tian
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | | | - Tianci Chai
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou
- Department of Anesthesiology, Xinyi People's Hospital, Xuzhou
| | | | | | | |
Collapse
|
17
|
Gupta R, Kumar P. Computational Analysis Indicates That PARP1 Acts as a Histone Deacetylases Interactor Sharing Common Lysine Residues for Acetylation, Ubiquitination, and SUMOylation in Alzheimer's and Parkinson's Disease. ACS OMEGA 2021; 6:5739-5753. [PMID: 33681613 PMCID: PMC7931403 DOI: 10.1021/acsomega.0c06168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/12/2021] [Indexed: 05/28/2023]
Abstract
Aim/Hypothesis : Lysine residues are known for the post-translational modifications (PTMs) such as acetylation, ubiquitination, and SUMOylation. In acetylation, histone deacetylase (HDAC) and its interactors cause transcriptional deregulation and cause mitochondrial dysfunction, apoptosis, inflammatory response, and cell-cycle impairment that cause brain homeostasis and neuronal cell death. Other regulatory PTMs involved in the pathogenesis of neurodegenerative diseases (NDDs) are ubiquitination and SUMOylation for the degradation of the misfolded proteins. Thus, we aim to investigate the potential acetylation/ubiquitination/SUMOylation crosstalk sites in the HDAC interactors, which cause NDDs. Furthermore, we aim to identify the influence of PTMs on the structural features of proteins and the impact of putative lysine mutation on disease susceptibility. Last, we aim to examine the impact of the putative mutation on acetylated lysine for ubiquitination and SUMOylation. Results : Herein, we integrate 1455 genes, 3094 genes, and 1940 genes related to HDAC interactors, Alzheimer's disease (AD), and Parkinson's disease (PD), respectively. Furthermore, the protein-protein interaction and PTM integrations from different databases identified 32 proteins that are associated with HDAC, AD, and PD with 1489 potential lysine-modified sites. HDAC interactors poly(ADP-ribose) polymerase 1 (PARP1), nucleophosmin (NPM1), and cyclin-dependent kinase 1 (CDK1) involved in the progression of NDDs and 64 and 75% of PTM sites in PARP1, NPM1, and CDK1 fall into coiled and ordered regions, respectively. Moreover, 15 putative lysine sites have been found in the crosstalk and K148, K249, K528, K637, K700, and K796 of PARP1 are crosstalk hotspots. Conclusion : The loss of acetylated hotspot sites results in the loss of ubiquitination and SUMOylation function on nearby sites, which is relatively higher when compared to the gain of function.
Collapse
|
18
|
Liu X, Qin J, Gao T, Li C, Chen X, Zeng K, Xu M, He B, Pan B, Xu X, Pan Y, Sun H, Xu T, Wang S. Analysis of METTL3 and METTL14 in hepatocellular carcinoma. Aging (Albany NY) 2020; 12:21638-21659. [PMID: 33159022 PMCID: PMC7695415 DOI: 10.18632/aging.103959] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/01/2020] [Indexed: 12/25/2022]
Abstract
N6-methyladenosine (m6A) RNA methylation is the most prevalent modification of messenger RNAs (mRNAs) and catalyzed by a multicomponent methyltransferase complex (MTC), among which methyltransferase-like 3 (METTL3) and METTL14 are two core molecules. However, METTL3 and METTL14 play opposite regulatory roles in hepatocellular carcinoma (HCC). Based on The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) database, we conducted a multi-omics analysis of METTL3 and METTL14 in HCC, including RNA-sequencing, m6ARIP-sequencing, and ribosome-sequencing profiles. We found that the expression and prognostic value of METTL3 and METTL14 are opposite in HCC. Besides, after METTL3 and METTL14 knockdown, most of the dysregulated mRNAs, signaling pathways and biological processes are distinct in HCC, which partly explains the contrary regulatory role of METTL3 and METTL14. Intriguingly, these mRNAs whose stability or translation efficiency are influenced by METTL3 or METTL14 in an m6A dependent manner, jointly regulate multiple signaling pathways and biological processes, which supports the cooperative role of METTL3 and METTL14 in catalyzing m6A modification. In conclusion, our study further clarified the contradictory role of METTL3 and METTL14 in HCC.
Collapse
Affiliation(s)
- Xiangxiang Liu
- School of Medicine, Southeast University, Nanjing 210096, Jiangsu, China
| | - Jian Qin
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Tianyi Gao
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Chenmeng Li
- School of Medicine, Southeast University, Nanjing 210096, Jiangsu, China
| | - Xiaoxiang Chen
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Kaixuan Zeng
- School of Medicine, Southeast University, Nanjing 210096, Jiangsu, China
| | - Mu Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Bangshun He
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Bei Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Xueni Xu
- School of Medicine, Southeast University, Nanjing 210096, Jiangsu, China
| | - Yuqin Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Huiling Sun
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Tao Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Shukui Wang
- School of Medicine, Southeast University, Nanjing 210096, Jiangsu, China.,Jiangsu Collaborative Innovation Center on Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211100, Jiangsu, China
| |
Collapse
|
19
|
Maddalena F, Condelli V, Matassa DS, Pacelli C, Scrima R, Lettini G, Li Bergolis V, Pietrafesa M, Crispo F, Piscazzi A, Storto G, Capitanio N, Esposito F, Landriscina M. TRAP1 enhances Warburg metabolism through modulation of PFK1 expression/activity and favors resistance to EGFR inhibitors in human colorectal carcinomas. Mol Oncol 2020; 14:3030-3047. [PMID: 33025742 PMCID: PMC7718945 DOI: 10.1002/1878-0261.12814] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 08/18/2020] [Accepted: 10/01/2020] [Indexed: 12/14/2022] Open
Abstract
Metabolic rewiring is a mechanism of adaptation to unfavorable environmental conditions and tumor progression. TRAP1 is an HSP90 molecular chaperone upregulated in human colorectal carcinomas (CRCs) and responsible for downregulation of oxidative phosphorylation (OXPHOS) and adaptation to metabolic stress. The mechanism by which TRAP1 regulates glycolytic metabolism and the relevance of this regulation in resistance to EGFR inhibitors were investigated in patient‐derived CRC spheres, human CRC cells, samples, and patients. A linear correlation was observed between TRAP1 levels and 18F‐fluoro‐2‐deoxy‐glucose (18F‐FDG) uptake upon PET scan or GLUT1 expression in human CRCs. Consistently, TRAP1 enhances GLUT1 expression, glucose uptake, and lactate production and downregulates OXPHOS in CRC patient‐derived spheroids and cell lines. Mechanistically, TRAP1 maximizes lactate production to balance low OXPHOS through the regulation of the glycolytic enzyme phosphofructokinase‐1 (PFK1); this depends on the interaction between TRAP1 and PFK1, which favors PFK1 glycolytic activity and prevents its ubiquitination/degradation. By contrast, TRAP1/PFK1 interaction is lost in conditions of enhanced OXPHOS, which results in loss of TRAP1 regulation of PFK1 activity and lactate production. Notably, TRAP1 regulation of glycolysis is involved in resistance of RAS‐wild‐type CRCs to EGFR monoclonals. Indeed, either TRAP1 upregulation or high glycolytic metabolism impairs cetuximab activity in vitro, whereas TRAP1 targeting and/or inhibition of glycolytic pathway enhances cell response to cetuximab. Finally, a linear correlation between 18F‐FDG PET uptake and poor response to cetuximab in first‐line therapy in human metastatic CRCs was observed. These results suggest that TRAP1 is a key determinant of CRC metabolic rewiring and favors resistance to EGFR inhibitors through regulation of glycolytic metabolism.
Collapse
Affiliation(s)
- Francesca Maddalena
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Valentina Condelli
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Danilo Swann Matassa
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Italy
| | - Consiglia Pacelli
- Department of Clinical and Experimental Medicine, University of Foggia, Italy
| | - Rosella Scrima
- Department of Clinical and Experimental Medicine, University of Foggia, Italy
| | - Giacomo Lettini
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Valeria Li Bergolis
- Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Italy
| | - Michele Pietrafesa
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Fabiana Crispo
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Annamaria Piscazzi
- Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Italy
| | - Giovanni Storto
- Nuclear Medicine Unit, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, Italy
| | - Franca Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Italy
| | - Matteo Landriscina
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy.,Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Italy
| |
Collapse
|
20
|
Lettini G, Condelli V, Pietrafesa M, Crispo F, Zoppoli P, Maddalena F, Laurenzana I, Sgambato A, Esposito F, Landriscina M. TRAP1 Regulates Wnt/β-Catenin Pathway through LRP5/6 Receptors Expression Modulation. Int J Mol Sci 2020; 21:E7526. [PMID: 33065966 PMCID: PMC7589514 DOI: 10.3390/ijms21207526] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/09/2020] [Accepted: 10/10/2020] [Indexed: 12/01/2022] Open
Abstract
Wnt/β-Catenin signaling is involved in embryonic development, regeneration, and cellular differentiation and is responsible for cancer stemness maintenance. The HSP90 molecular chaperone TRAP1 is upregulated in 60-70% of human colorectal carcinomas (CRCs) and favors stem cells maintenance, modulating the Wnt/β-Catenin pathway and preventing β-Catenin phosphorylation/degradation. The role of TRAP1 in the regulation of Wnt/β-Catenin signaling was further investigated in human CRC cell lines, patient-derived spheroids, and CRC specimens. TRAP1 relevance in the activation of Wnt/β-Catenin signaling was highlighted by a TCF/LEF Cignal Reporter Assay in Wnt-off HEK293T and CRC HCT116 cell lines. Of note, this regulation occurs through the modulation of Wnt ligand receptors LRP5 and LRP6 that are both downregulated in TRAP1-silenced cell lines. However, while LRP5 mRNA is significantly downregulated upon TRAP1 silencing, LRP6 mRNA is unchanged, suggesting independent mechanisms of regulation by TRAP1. Indeed, LRP5 is regulated upon promoter methylation in CRC cell lines and human CRCs, whereas LRP6 is controlled at post-translational level by protein ubiquitination/degradation. Consistently, human CRCs with high TRAP1 expression are characterized by the co-upregulation of active β-Catenin, LRP5 and LRP6. Altogether, these data suggest that Wnt/β-Catenin signaling is modulated at multiple levels by TRAP1.
Collapse
Affiliation(s)
- Giacomo Lettini
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, PZ, Italy; (G.L.); (V.C.); (M.P.); (F.C.); (P.Z.); (F.M.); (I.L.); (A.S.)
| | - Valentina Condelli
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, PZ, Italy; (G.L.); (V.C.); (M.P.); (F.C.); (P.Z.); (F.M.); (I.L.); (A.S.)
| | - Michele Pietrafesa
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, PZ, Italy; (G.L.); (V.C.); (M.P.); (F.C.); (P.Z.); (F.M.); (I.L.); (A.S.)
| | - Fabiana Crispo
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, PZ, Italy; (G.L.); (V.C.); (M.P.); (F.C.); (P.Z.); (F.M.); (I.L.); (A.S.)
| | - Pietro Zoppoli
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, PZ, Italy; (G.L.); (V.C.); (M.P.); (F.C.); (P.Z.); (F.M.); (I.L.); (A.S.)
| | - Francesca Maddalena
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, PZ, Italy; (G.L.); (V.C.); (M.P.); (F.C.); (P.Z.); (F.M.); (I.L.); (A.S.)
| | - Ilaria Laurenzana
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, PZ, Italy; (G.L.); (V.C.); (M.P.); (F.C.); (P.Z.); (F.M.); (I.L.); (A.S.)
| | - Alessandro Sgambato
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, PZ, Italy; (G.L.); (V.C.); (M.P.); (F.C.); (P.Z.); (F.M.); (I.L.); (A.S.)
| | - Franca Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Matteo Landriscina
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, PZ, Italy; (G.L.); (V.C.); (M.P.); (F.C.); (P.Z.); (F.M.); (I.L.); (A.S.)
- Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy
| |
Collapse
|
21
|
Identification of candidate aberrantly methylated and differentially expressed genes in Esophageal squamous cell carcinoma. Sci Rep 2020; 10:9735. [PMID: 32546690 PMCID: PMC7297810 DOI: 10.1038/s41598-020-66847-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 05/28/2020] [Indexed: 12/11/2022] Open
Abstract
Aberrant methylated genes (DMGs) play an important role in the etiology and pathogenesis of esophageal squamous cell carcinoma (ESCC). In this study, we aimed to integrate three cohorts profile datasets to ascertain aberrant methylated-differentially expressed genes and pathways associated with ESCC by comprehensive bioinformatics analysis. We downloaded data of gene expression microarrays (GSE20347, GSE38129) and gene methylation microarrays (GSE52826) from the Gene Expression Omnibus (GEO) database. Aberrantly differentially expressed genes (DEGs) were obtained by GEO2R tool. The David database was then used to perform Gene ontology (GO) analysis and Kyoto Encyclopedia of Gene and Genome pathway enrichment analyses on selected genes. STRING and Cytoscape software were used to construct a protein-protein interaction (PPI) network, then the modules in the PPI networks were analyzed with MCODE and the hub genes chose from the PPI networks were verified by Oncomine and TCGA database. In total, 291 hypomethylation-high expression genes and 168 hypermethylation-low expression genes were identified at the screening step, and finally found six mostly changed hub genes including KIF14, CDK1, AURKA, LCN2, TGM1, and DSG1. Pathway analysis indicated that aberrantly methylated DEGs mainly associated with the P13K-AKT signaling, cAMP signaling and cell cycle process. After validation in multiple databases, most hub genes remained significant. Patients with high expression of AURKA were associated with shorter overall survival. To summarize, we have identified six feasible aberrant methylated-differentially expressed genes and pathways in ESCC by bioinformatics analysis, potentially providing valuable information for the molecular mechanisms of ESCC. Our data combined the analysis of gene expression profiling microarrays and gene methylation profiling microarrays, simultaneously, and in this way, it can shed a light for screening and diagnosis of ESCC in future.
Collapse
|
22
|
New insights into molecular chaperone TRAP1 as a feasible target for future cancer treatments. Life Sci 2020; 254:117737. [PMID: 32376268 DOI: 10.1016/j.lfs.2020.117737] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/07/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023]
Abstract
Tumor necrosis factor receptor-associated protein 1 (TRAP1), a molecular chaperone, is a major member of the mitochondrial heat shock protein 90 (Hsp90) family. Studies have shown that TRAP1 can prevent hypoxia-induced damage to cardiomyocytes, maintain cardiomyocytes viability and mitochondrial membrane potential, and protect cardiomyocytes. In addition, it can also protect astrocytes from ischemic damage in vitro. In recent years, there have been many new discoveries in tumors. The abnormal expression of TRAP1 is closely related to the occurrence and development of various tumors. TRAP1 protein seems to be a central regulatory protein, involved in the activation of various oncogenic proteins and signaling pathways, and has a balanced function at tumor transformation and the intersection of different metabolic processes. Targeting its chaperone activity and molecular interactions can destroy the metabolism and survival adaptability of tumor cells, paving the way for the development of highly selective mitochondrial anti-tumor drugs. Moreover, the combination of TRAP1 inhibition and current traditional cancer therapies has shown promising applications. These findings have important implications for the diagnosis and treatment of tumors. Therefore, we reviewed the recently identified functions of the molecular chaperone TRAP1 in cancer development and progression, as well as the discovery and recent advances in selective TRAP1 inhibitors as anticancer drug therapies, opening up new attractive prospects for exploring strategies for targeting TRAP1 as a tumor cell target.
Collapse
|
23
|
Lettini G, Pietrafesa M, Lepore S, Maddalena F, Crispo F, Sgambato A, Esposito F, Landriscina M. Heat shock proteins in thyroid malignancies: Potential therapeutic targets for poorly-differentiated and anaplastic tumours? Mol Cell Endocrinol 2020; 502:110676. [PMID: 31812782 DOI: 10.1016/j.mce.2019.110676] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/03/2019] [Accepted: 12/03/2019] [Indexed: 12/31/2022]
Abstract
Thyroid cancer is the most common endocrine malignancy, with well-differentiated subtypes characterized by an excellent prognosis due to their optimal sensitivity to standard therapies whereas poorly differentiated and anaplastic tumours by chemo/radio-resistance and unfavourable outcome. Heat Shock Proteins (HSPs) are molecular chaperones overexpressed in thyroid malignancies and involved in crucial functions responsible for thyroid carcinogenesis, as protection from apoptosis, drug resistance and cell migration. Thus, HSPs inhibitors have been proposed as novel therapeutic agents in thyroid cancer to revert molecular mechanisms of tumour progression. In this review, we report an overview on the biological role of HSPs, and specifically HSP90s, in thyroid cancer and their potential involvement as biomarkers. We discuss the rationale to evaluate HSPs inhibitors as innovative anticancer agents in specific subtypes of thyroid cancer characterized by poor response to therapies with the objective to target single family chaperones to reduce, simultaneously, the expression/stability of multiple client proteins.
Collapse
Affiliation(s)
- Giacomo Lettini
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Michele Pietrafesa
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Silvia Lepore
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Francesca Maddalena
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Fabiana Crispo
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Alessandro Sgambato
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Franca Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Napoli Federico II, Naples, Italy.
| | - Matteo Landriscina
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy; Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| |
Collapse
|
24
|
Gene Copy Number and Post-Transductional Mechanisms Regulate TRAP1 Expression in Human Colorectal Carcinomas. Int J Mol Sci 2019; 21:ijms21010145. [PMID: 31878280 PMCID: PMC6981705 DOI: 10.3390/ijms21010145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/17/2019] [Accepted: 12/20/2019] [Indexed: 12/29/2022] Open
Abstract
Tumor Necrosis Factor Receptor-Associated Protein 1 (TRAP1) is a heat shock protein 90 (HSP90) molecular chaperone overexpressed in 60-70% human colorectal carcinomas (CRCs) and the co-upregulation of TRAP1 and associated 6-related proteins identifies metastatic CRCs with poor prognosis. Since the molecular mechanisms responsible for TRAP1 regulation are still unknown, the significance of TRAP1 gene copy number (CN) and the role of post-transductional protein modifications were addressed. TRAP1 gene aneuploidy accounted for 34.5% of cases in a cohort of 58 human CRCs and TRAP1 CN correlated with its mRNA and protein expression, suggesting that transcriptional mechanisms are responsible for TRAP1 upregulation. Furthermore, the analysis of the National Cancer Institute's Clinical Proteomic Tumor Analysis Consortium/The Cancer Genome Atlas (CPTAC/TCGA) CRC database showed that TRAP1 polysomy significantly correlates with lymph node involvement. However, a subgroup of tumors showed TRAP1 protein levels independent from its CN. Of note, a direct correlation was observed between TRAP1 protein levels and the expression of S-nitrosoglutathione reductase (GSNOR), a denitrosylase involved in the regulation of protein S-nitrosylation. Furthermore, CRC cell lines exposed to hypoxia or dichloroacetate treatment showed the downregulation of TRAP1 upon GSNOR silencing and this resulted in increased TRAP1 mono/polyubiquitination. These data suggest that transcriptional and post-transductional mechanisms account for TRAP1 expression in human CRCs and GSNOR protects TRAP1 from S-nitrosylation and consequent proteasome degradation mostly in conditions of stress.
Collapse
|
25
|
Zou YF, Rong YM, Tan YX, Xiao J, Yu ZL, Chen YF, Ke J, Li CH, Chen X, Wu XJ, Lan P, Lin XT, Gao F. A signature of hypoxia-related factors reveals functional dysregulation and robustly predicts clinical outcomes in stage I/II colorectal cancer patients. Cancer Cell Int 2019; 19:243. [PMID: 31572060 PMCID: PMC6757395 DOI: 10.1186/s12935-019-0964-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/13/2019] [Indexed: 12/27/2022] Open
Abstract
Background The hypoxic tumor microenvironment accelerates the invasion and migration of colorectal cancer (CRC) cells. The aim of this study was to develop and validate a hypoxia gene signature for predicting the outcome in stage I/II CRC patients that have limited therapeutic options. Methods The hypoxic gene signature (HGS) was constructed using transcriptomic data of 309 CRC patients with complete clinical information from the CIT microarray dataset. A total of 1877 CRC patients with complete prognostic information in six independent datasets were divided into a training cohort and two validation cohorts. Univariate and multivariate analyses were conducted to evaluate the prognostic value of HGS. Results The HGS consisted of 14 genes, and demarcated the CRC patients into the high- and low-risk groups. In all three cohorts, patients in the high-risk group had significantly worse disease free survival (DFS) compared with those in the low risk group (training cohort—HR = 4.35, 95% CI 2.30–8.23, P < 0.001; TCGA cohort—HR = 2.14, 95% CI 1.09–4.21, P = 0.024; meta-validation cohort—HR = 1.91, 95% CI 1.08–3.39, P = 0.024). Compared to Oncotype DX, HGS showed superior predictive outcome in the training cohort (C-index, 0.80 vs 0.65) and the validation cohort (C-index, 0.70 vs 0.61). Pathway analysis of the high- and low-HGS groups showed significant differences in the expression of genes involved in mTROC1, G2-M, mitosis, oxidative phosphorylation, MYC and PI3K–AKT–mTOR pathways (P < 0.005). Conclusion Hypoxic gene signature is a satisfactory prognostic model for early stage CRC patients, and the exact biological mechanism needs to be validated further.
Collapse
Affiliation(s)
- Yi-Feng Zou
- 1Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong China.,4Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, 510655 Guangdong China
| | - Yu-Ming Rong
- 5Department of VIP Region, Cancer Center of Sun Yat-sen University, Guangzhou, Guangdong China
| | - Ying-Xin Tan
- 1Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong China.,4Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, 510655 Guangdong China
| | - Jian Xiao
- 2Department of Medical Oncology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong China.,4Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, 510655 Guangdong China
| | - Zhao-Liang Yu
- 1Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong China.,4Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, 510655 Guangdong China
| | - Yu-Feng Chen
- 1Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong China.,4Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, 510655 Guangdong China
| | - Jia Ke
- 1Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong China.,4Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, 510655 Guangdong China
| | - Cheng-Hang Li
- 1Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong China.,4Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, 510655 Guangdong China
| | - Xi Chen
- 1Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong China.,4Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, 510655 Guangdong China
| | - Xiao-Jian Wu
- 1Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong China.,4Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, 510655 Guangdong China
| | - Ping Lan
- 1Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong China.,4Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, 510655 Guangdong China
| | - Xu-Tao Lin
- 1Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong China.,3Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong China.,4Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, 510655 Guangdong China
| | - Feng Gao
- 1Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong China.,4Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, 510655 Guangdong China
| |
Collapse
|
26
|
Liu LL, Zhu JM, Yu XN, Zhu HR, Shi X, Bilegsaikhan E, Guo HY, Wu J, Shen XZ. UBE2T promotes proliferation via G2/M checkpoint in hepatocellular carcinoma. Cancer Manag Res 2019; 11:8359-8370. [PMID: 31571992 PMCID: PMC6750879 DOI: 10.2147/cmar.s202631] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 08/19/2019] [Indexed: 12/15/2022] Open
Abstract
Background Growing evidence suggests that the ubiquitin-proteasome system is involved in the pathogenesis and recurrence of hepatocellular carcinoma (HCC); yet, little is known about the role of ubiquitin-conjugating enzyme E2T (UBE2T) in HCC. Materials and methods UBE2T levels were detected in HCC tissues and hepatoma cell lines using quantitative reserve transcriptase-polymerase chain reaction and Western blot analysis. Next, the changes of phenotypes after UBE2T knockdown or overexpression were evaluated using in vitro methods. Finally, the mechanism of UBE2T in HCC was tested using ex vivo and in vivo methods. Results In the present study, we reported that UBE2T mRNA and protein levels were significantly upregulated in HCC tissues compared to adjacent non-tumor tissues. Additionally, suppression of UBE2T expression inhibited proliferation, colony formation, tumorigenesis, migration, and invasion of hepatoma cells, whereas UBE2T overexpression led to the opposite outcomes. Moreover, suppression of UBE2T expression resulted in an increase in G2/M phase and a decrease in the percentage of cells in G1 phase, which indicated a cell cycle arrest at the G2/M phase. In contrast, the percentage of cells in G2/M phase decreased following UBE2T overexpression. Further study indicated that UBE2T regulated the G2/M transition by modulating cyclin B1 and cyclin-dependent kinase 1. Conclusion Taken together, the findings of the present study uncover biological functions of UBE2T in hepatoma cells, and delineate preliminary molecular mechanisms of UBE2T in modulating HCC development and progression.
Collapse
Affiliation(s)
- Li-Li Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China.,Shanghai Institute of Liver Diseases, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Ji-Min Zhu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China.,Shanghai Institute of Liver Diseases, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Xiang-Nan Yu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China.,Shanghai Institute of Liver Diseases, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Hai-Rong Zhu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China.,Shanghai Institute of Liver Diseases, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Xuan Shi
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China.,Shanghai Institute of Liver Diseases, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Enkhnaran Bilegsaikhan
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China.,Shanghai Institute of Liver Diseases, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Hong-Ying Guo
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China.,Shanghai Institute of Liver Diseases, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Jian Wu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China.,Shanghai Institute of Liver Diseases, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Department of Medical Microbiology and Parasitology, Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences, Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Xi-Zhong Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China.,Shanghai Institute of Liver Diseases, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Department of Medical Microbiology and Parasitology, Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences, Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| |
Collapse
|
27
|
Lettini G, Lepore S, Crispo F, Sisinni L, Esposito F, Landriscina M. Heat shock proteins in cancer stem cell maintenance: A potential therapeutic target? Histol Histopathol 2019; 35:25-37. [PMID: 31322279 DOI: 10.14670/hh-18-153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer stem cells (CSCs) are a subpopulation of tumor cells with unlimited self-renewal capability, multilineage differentiation potential and long-term tumor repopulation capacity. CSCs reside in anatomically distinct regions within the tumor microenvironment, called niches, and this favors the maintenance of CSC properties and preserves their phenotypic plasticity. Indeed, CSCs are characterized by a flexible state based on their capacity to interconvert between a differentiated and a stem-like phenotype, and this depends on the activation of adaptive mechanisms in response to different environmental conditions. Heat Shock Proteins (HSPs) are molecular chaperones, upregulated upon cell exposure to several stress conditions and are responsible for normal maturation, localization and activity of intra and extracellular proteins. Noteworthy, HSPs play a central role in several cellular processes involved in tumor initiation and progression (i.e. cell viability, resistance to apoptosis, stress conditions and drug therapy, EMT, bioenergetics, invasiveness, metastasis formation) and, thus, are widely considered potential molecular targets. Furthermore, much evidence suggests a key regulatory function for HSPs in CSC maintenance and their upregulation has been proposed as a mechanism used by CSCs to adapt to unfavorable environmental conditions, such as nutrient deprivation, hypoxia, inflammation. This review discusses the relevance of HSPs in CSC biology, highlighting their role as novel potential molecular targets to develop anticancer strategies aimed at CSC targeting.
Collapse
Affiliation(s)
- Giacomo Lettini
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Silvia Lepore
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Fabiana Crispo
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Lorenza Sisinni
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Franca Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy.
| | - Matteo Landriscina
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy.,Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| |
Collapse
|
28
|
Condelli V, Crispo F, Pietrafesa M, Lettini G, Matassa DS, Esposito F, Landriscina M, Maddalena F. HSP90 Molecular Chaperones, Metabolic Rewiring, and Epigenetics: Impact on Tumor Progression and Perspective for Anticancer Therapy. Cells 2019; 8:cells8060532. [PMID: 31163702 PMCID: PMC6627532 DOI: 10.3390/cells8060532] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 12/31/2022] Open
Abstract
Heat shock protein 90 (HSP90) molecular chaperones are a family of ubiquitous proteins participating in several cellular functions through the regulation of folding and/or assembly of large multiprotein complexes and client proteins. Thus, HSP90s chaperones are, directly or indirectly, master regulators of a variety of cellular processes, such as adaptation to stress, cell proliferation, motility, angiogenesis, and signal transduction. In recent years, it has been proposed that HSP90s play a crucial role in carcinogenesis as regulators of genotype-to-phenotype interplay. Indeed, HSP90 chaperones control metabolic rewiring, a hallmark of cancer cells, and influence the transcription of several of the key-genes responsible for tumorigenesis and cancer progression, through either direct binding to chromatin or through the quality control of transcription factors and epigenetic effectors. In this review, we will revise evidence suggesting how this interplay between epigenetics and metabolism may affect oncogenesis. We will examine the effect of metabolic rewiring on the accumulation of specific metabolites, and the changes in the availability of epigenetic co-factors and how this process can be controlled by HSP90 molecular chaperones. Understanding deeply the relationship between epigenetic and metabolism could disclose novel therapeutic scenarios that may lead to improvements in cancer treatment.
Collapse
Affiliation(s)
- Valentina Condelli
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, PZ, Italy.
| | - Fabiana Crispo
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, PZ, Italy.
| | - Michele Pietrafesa
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, PZ, Italy.
| | - Giacomo Lettini
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, PZ, Italy.
| | - Danilo Swann Matassa
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy.
| | - Franca Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy.
| | - Matteo Landriscina
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, PZ, Italy.
- Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy.
| | - Francesca Maddalena
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, PZ, Italy.
| |
Collapse
|
29
|
Liu K, Kang M, Li J, Qin W, Wang R. Prognostic value of the mRNA expression of members of the HSP90 family in non-small cell lung cancer. Exp Ther Med 2019; 17:2657-2665. [PMID: 30930968 PMCID: PMC6425268 DOI: 10.3892/etm.2019.7228] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 12/21/2018] [Indexed: 12/15/2022] Open
Abstract
The aim of the present study was to investigate the potential prognostic value of members of the heat shock protein (HSP)90 family in non-small cell lung cancer (NSCLC) patients. The mRNA expression profiles of 1,926 NSCLC patients, which was available from the Kaplan-Meier plotter database, were included in the study. High expression of HSP90AA1 mRNA was significantly associated with a poorer rate of overall survival (OS) for all NSCLC patients [hazard ratio (HR), 1.21; 95% confidence interval (CI): 1.06–1.37; P=0.004], as well as for patients with adenocarcinoma (ADE; HR, 1.3; 95% CI: 1.02–1.65; P=0.034), but no significant correlation was identified for squamous cell carcinoma (SCC) patients (HR, 1.08; 95% CI: 0.85–1.38; P=0.51). High expression of HSP90AB1 and HSP90B1 mRNA was significantly associated with poorer rates of OS in lung SCC and ADE patients combined, as well as in lung ADE patients alone. By contrast, high expression of tumor necrosis factor receptor-associated protein 1 (TRAP1) mRNA was significantly associated with improved OS rates in all NSCLC patients combined (HR, 0.88; 95% CI: 0.77–0.99; P=0.041), as well as ADE patients. In stratified survival analysis, a high expression of HSP90AA1, HSP90AB1 and HSP90B1 predicted poor prognosis in stage I NSLCC patients, suggesting that these genes may serve as stage-independent prognostic indicators. As an elevated expression of HSP90AA1, HSP90AB1, HSP90B1 and TRAP1 was associated with poorer OS outcomes in patients with NSCLC, these HSP90 members may be potential prognostic biomarkers and drug targets for the treatment of NSCLC.
Collapse
Affiliation(s)
- Kang Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Min Kang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jixi Li
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Wen Qin
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Rensheng Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
30
|
Spagnoletti G, Li Bergolis V, Piscazzi A, Giannelli F, Condelli V, Sisinni L, Bove G, Storto G, Landriscina M. Cyclin-dependent kinase 1 targeting improves sensitivity to radiation in BRAF V600E colorectal carcinoma cells. Tumour Biol 2018; 40:1010428318770957. [PMID: 29663854 DOI: 10.1177/1010428318770957] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Preoperative chemoradiation is currently the standard of care in locally advanced rectal carcinoma, even though a subset of rectal tumors does not achieve major clinically meaningful responses upon neoadjuvant chemoradiation. At present, no molecular biomarkers are available to predict response to neoadjuvant chemoradiation and select resistant tumors willing more intense therapeutic strategies. Thus, BRAF mutational status was investigated for its role in favoring resistance to radiation in colorectal carcinoma cell lines and cyclin-dependent kinase 1 as a target to improve radiosensitivity in BRAF V600E colorectal tumor cells. METHODS Colony-forming assay and apoptotic rates were evaluated to compare the sensitivity of different colon carcinoma cell lines to ionizing radiation and their radiosensitivity upon exposure to BRAF and/or cyclin-dependent kinase 1 inhibitory/silencing strategies. Cyclin-dependent kinase 1 expression/subcellular distribution was studied by immunoblot analysis. RESULTS Colon carcinoma BRAF V600E HT29 cells exhibited poor response to radiation compared to BRAF wild-type COLO320 and HCT116 cells. Interestingly, neither radiosensitizing doses of 5-fluoruracil nor BRAF inhibition/silencing significantly improved radiosensitivity in HT29 cells. Of note, poor response to radiation correlated with upregulation/relocation of cyclin-dependent kinase 1 in mitochondria. Consistently, cyclin-dependent kinase 1 inhibition/silencing as well as its targeting, through inhibition of HSP90 quality control pathway, significantly inhibited the clonogenic ability and increased apoptotic rates in HT29 cells upon exposure to radiation. CONCLUSION These data suggest that BRAF V600E colorectal carcinoma cells are poorly responsive to radiation, and cyclin-dependent kinase 1 represents a target to improve radiosensitivity in BRAF V600E colorectal tumor cells.
Collapse
Affiliation(s)
- Girolamo Spagnoletti
- 1 Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.,2 Radiotherapy Unit, Ospedali Riuniti, Foggia, Italy
| | - Valeria Li Bergolis
- 1 Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Annamaria Piscazzi
- 1 Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Francesca Giannelli
- 1 Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Valentina Condelli
- 3 Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Pz, Italy
| | - Lorenza Sisinni
- 3 Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Pz, Italy
| | - Giuseppe Bove
- 2 Radiotherapy Unit, Ospedali Riuniti, Foggia, Italy
| | - Giovanni Storto
- 4 Nuclear Medicine Unit, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Pz, Italy
| | - Matteo Landriscina
- 1 Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.,3 Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Pz, Italy
| |
Collapse
|
31
|
Matassa DS, Agliarulo I, Avolio R, Landriscina M, Esposito F. TRAP1 Regulation of Cancer Metabolism: Dual Role as Oncogene or Tumor Suppressor. Genes (Basel) 2018; 9:genes9040195. [PMID: 29621137 PMCID: PMC5924537 DOI: 10.3390/genes9040195] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 03/28/2018] [Accepted: 03/28/2018] [Indexed: 12/20/2022] Open
Abstract
Metabolic reprogramming is an important issue in tumor biology. An unexpected inter- and intra-tumor metabolic heterogeneity has been strictly correlated to tumor outcome. Tumor Necrosis Factor Receptor-Associated Protein 1 (TRAP1) is a molecular chaperone involved in the regulation of energetic metabolism in cancer cells. This protein is highly expressed in several cancers, such as glioblastoma, colon, breast, prostate and lung cancers and is often associated with drug resistance. However, TRAP1 is also downregulated in specific tumors, such as ovarian, bladder and renal cancers, where its lower expression is correlated with the worst prognoses and chemoresistance. TRAP1 is the only mitochondrial member of the Heat Shock Protein 90 (HSP90) family that directly interacts with respiratory complexes, contributing to their stability and activity but it is still unclear if such interactions lead to reduced or increased respiratory capacity. The role of TRAP1 is to enhance or suppress oxidative phosphorylation; the effects of such regulation on tumor development and progression are controversial. These observations encourage the study of the mechanisms responsible for the dualist role of TRAP1 as an oncogene or oncosuppressor in specific tumor types. In this review, TRAP1 puzzling functions were recapitulated with a special focus on the correlation between metabolic reprogramming and tumor outcome. We wanted to investigate whether metabolism-targeting drugs can efficiently interfere with tumor progression and whether they might be combined with chemotherapeutics or molecular-targeted agents to counteract drug resistance and reduce therapeutic failure.
Collapse
Affiliation(s)
- Danilo Swann Matassa
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy.
| | - Ilenia Agliarulo
- Institute of Protein Biochemistry (IBP), National Research Council, 80131 Naples, Italy.
| | - Rosario Avolio
- Gene Regulation, Stem Cells and Cancer Programme, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain.
| | - Matteo Landriscina
- Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, 7100 Foggia, Italy.
- Laboratory of Pre-clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy.
| | - Franca Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy.
| |
Collapse
|
32
|
Lettini G, Maddalena F, Sisinni L, Condelli V, Matassa DS, Costi MP, Simoni D, Esposito F, Landriscina M. TRAP1: a viable therapeutic target for future cancer treatments? Expert Opin Ther Targets 2017; 21:805-815. [PMID: 28664757 DOI: 10.1080/14728222.2017.1349755] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION HSP90 molecular chaperones (i.e., HSP90α, HSP90β, GRP94 and TRAP1) are potential therapeutic targets to design novel anticancer agents. However, despite numerous designed HSP90 inhibitors, most of them have failed due to unfavorable toxicity profiles and lack of specificity toward different HSP90 paralogs. Indeed, a major limitation in this field is the high structural homology between different HSP90 chaperones, which significantly limits our capacity to design paralog-specific inhibitors. Area covered: This review examines the relevance of TRAP1 in tumor development and progression, with an emphasis on its oncogenic/oncosuppressive role in specific human malignancies and its multifaceted and context-dependent functions in cancer cells. Herein, we discuss the rationale for considering TRAP1 as a potential molecular target and the strategies used to date, to achieve its compartmentalized inhibition directly in mitochondria. Expert opinion: TRAP1 targeting may represent a promising strategy for cancer therapy, based on the increasing and compelling evidence supporting TRAP1 involvement in human carcinogenesis. However, considering the complexity of TRAP1 biology, future strategies of drug discovery need to improve selectivity and specificity toward TRAP1 respect to other HSP90 paralogs. The characterization of specific human malignancies suitable for TRAP1 targeting is also mandatory.
Collapse
Affiliation(s)
- Giacomo Lettini
- a Laboratory of Pre-Clinical and Translational Research , IRCCS, Referral Cancer Center of Basilicata , Rionero in Vulture , Italy
| | - Francesca Maddalena
- a Laboratory of Pre-Clinical and Translational Research , IRCCS, Referral Cancer Center of Basilicata , Rionero in Vulture , Italy
| | - Lorenza Sisinni
- a Laboratory of Pre-Clinical and Translational Research , IRCCS, Referral Cancer Center of Basilicata , Rionero in Vulture , Italy
| | - Valentina Condelli
- a Laboratory of Pre-Clinical and Translational Research , IRCCS, Referral Cancer Center of Basilicata , Rionero in Vulture , Italy
| | - Danilo Swann Matassa
- b Department of Molecular Medicine and Medical Biotechnology , University of Naples Federico II , Napoli , Italy
| | - Maria Paola Costi
- c Department of Life Sciences , University of Modena and Reggio Emilia , Modena , Italy
| | - Daniele Simoni
- d Department of Chemical and Pharmaceutical Sciences , University of Ferrara , Ferrara , Italy
| | - Franca Esposito
- b Department of Molecular Medicine and Medical Biotechnology , University of Naples Federico II , Napoli , Italy
| | - Matteo Landriscina
- a Laboratory of Pre-Clinical and Translational Research , IRCCS, Referral Cancer Center of Basilicata , Rionero in Vulture , Italy.,e Medical Oncology Unit, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| |
Collapse
|