1
|
Alsofyani AA, Nedjadi T. Gelsolin, an Actin-Binding Protein: Bioinformatic Analysis and Functional Significance in Urothelial Bladder Carcinoma. Int J Mol Sci 2023; 24:15763. [PMID: 37958747 PMCID: PMC10647509 DOI: 10.3390/ijms242115763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 09/17/2023] [Accepted: 09/18/2023] [Indexed: 11/15/2023] Open
Abstract
The involvement of the actin-regulatory protein, gelsolin (GSN), in neoplastic transformation has been reported in different cancers including bladder cancer. However, the exact mechanism by which GSN influences bladder cancer development is not well understood. Here, we sought to reveal the functional significance of GSN in bladder cancer by undertaking a comprehensive bioinformatic analysis of TCGA datasets and through the assessment of multiple biological functions. GSN expression was knocked down in bladder cancer cell lines with two siRNA isoforms targeting GSN. Proliferation, migration, cell cycle and apoptosis assays were carried out. GSN expression, enrichment analysis, protein-protein interaction and immune infiltration analysis were verified through online TCGA tools. The data indicated that GSN expression is associated with bladder cancer proliferation, migration and enhanced cell apoptosis through regulation of NF-κB expression. GSN expression correlated with various inflammatory cells and may influence the immunity of the tumor microenvironment. Computational analysis identified several interacting partners which are associated with cancer progression and patient outcome. The present results demonstrate that GSN plays an important role in bladder cancer pathogenesis and may serve as a potential biomarker and therapeutic target for cancer therapy.
Collapse
Affiliation(s)
| | - Taoufik Nedjadi
- King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Jeddah 21423, Saudi Arabia;
| |
Collapse
|
2
|
Zhang Q, Wen XH, Tang SL, Zhao ZW, Tang CK. Role and therapeutic potential of gelsolin in atherosclerosis. J Mol Cell Cardiol 2023; 178:59-67. [PMID: 36967105 DOI: 10.1016/j.yjmcc.2023.03.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 04/08/2023]
Abstract
Atherosclerosis is the major pathophysiological basis of a variety of cardiovascular diseases and has been recognized as a lipid-driven chronic inflammatory disease. Gelsolin (GSN) is a member of the GSN family. The main function of GSN is to cut and seal actin filaments to regulate the cytoskeleton and participate in a variety of biological functions, such as cell movement, morphological changes, metabolism, apoptosis and phagocytosis. Recently, more and more evidences have demonstrated that GSN is Closely related to atherosclerosis, involving lipid metabolism, inflammation, cell proliferation, migration and thrombosis. This article reviews the role of GSN in atherosclerosis from inflammation, apoptosis, angiogenesis and thrombosis.
Collapse
Affiliation(s)
- Qiang Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Department of Intensive Care Unit, the First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiao-Hui Wen
- School of Nursing, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Shi-Lin Tang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Department of Intensive Care Unit, the First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhen-Wang Zhao
- School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, Hubei 441053, China.
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Department of Intensive Care Unit, the First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
3
|
The p53 and Calcium Regulated Actin Rearrangement in Model Cells. Int J Mol Sci 2022; 23:ijms23169078. [PMID: 36012344 PMCID: PMC9408879 DOI: 10.3390/ijms23169078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/04/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
Long-term cellular stress maintains high intracellular Ca2+ concentrations which ultimately initiates apoptosis. Our interest is focused on how the gelsolin (GSN) and junctional mediating and regulating Y protein (JMY) play important roles in stress response. Both of these proteins can bind p53 and actin. We investigated using in vitro fluorescence spectroscopy and found that the p53 competes with actin in GSN to inhibit p53–JMY complex formation. A high Ca2+ level initializes p53 dimerization; the dimer competes with actin on JMY, which can lead to p53–JMY cotransport into the nucleus. Here we investigated how the motility and division rate of HeLa cells changes due to low-voltage electroporation of GSN or JMY in scratching assays. We revealed that JMY inhibits their motion, but that it can accelerate the cell division. GSN treatment slows down cell division but does not affect cell motility. HeLa cells fully recovered the gap 20 h after the electroporation with JMY and then started to release from the glass slides. Taken together, our in vitro results indicate that GSN and JMY may play an important role in the cellular stress response.
Collapse
|
4
|
Quaresma MC, Botelho HM, Pankonien I, Rodrigues CS, Pinto MC, Costa PR, Duarte A, Amaral MD. Exploring YAP1-centered networks linking dysfunctional CFTR to epithelial-mesenchymal transition. Life Sci Alliance 2022; 5:5/9/e202101326. [PMID: 35500936 PMCID: PMC9060002 DOI: 10.26508/lsa.202101326] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/20/2022] [Accepted: 04/20/2022] [Indexed: 12/21/2022] Open
Abstract
In this work, a systems biology approach identifies potentially dysregulated EMT signaling in CF (including the Hippo, Wnt, TGF-β, p53, and MYC pathways), integrated by YAP1 and TEAD4. Mutations in the CFTR anion channel cause cystic fibrosis (CF) and have also been related to higher cancer incidence. Previously we proposed that this is linked to an emerging role of functional CFTR in protecting against epithelial–mesenchymal transition (EMT). However, the pathways bridging dysfunctional CFTR to EMT remain elusive. Here, we applied systems biology to address this question. Our data show that YAP1 is aberrantly active in the presence of mutant CFTR, interacting with F508del, but not with wt-CFTR, and that YAP1 knockdown rescues F508del-CFTR processing and function. Subsequent analysis of YAP1 interactors and roles in cells expressing either wt- or F508del-CFTR reveal that YAP1 is an important mediator of the fibrotic/EMT processes in CF. Alongside, five main pathways emerge here as key in linking mutant CFTR to EMT, namely, (1) the Hippo pathway; (2) the Wnt pathway; (3) the TGFβ pathway; (4) the p53 pathway; and (5) MYC signaling. Several potential hub proteins which mediate the crosstalk among these pathways were also identified, appearing as potential therapeutic targets for both CF and cancer.
Collapse
Affiliation(s)
- Margarida C Quaresma
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| | - Hugo M Botelho
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| | - Ines Pankonien
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| | - Cláudia S Rodrigues
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| | - Madalena C Pinto
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| | - Pau R Costa
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| | - Aires Duarte
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| | - Margarida D Amaral
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| |
Collapse
|
5
|
Illescas M, Peñas A, Arenas J, Martín MA, Ugalde C. Regulation of Mitochondrial Function by the Actin Cytoskeleton. Front Cell Dev Biol 2022; 9:795838. [PMID: 34993202 PMCID: PMC8725978 DOI: 10.3389/fcell.2021.795838] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/03/2021] [Indexed: 12/13/2022] Open
Abstract
The regulatory role of actin cytoskeleton on mitochondrial function is a growing research field, but the underlying molecular mechanisms remain poorly understood. Specific actin-binding proteins (ABPs), such as Gelsolin, have also been shown to participate in the pathophysiology of mitochondrial OXPHOS disorders through yet to be defined mechanisms. In this mini-review, we will summarize the experimental evidence supporting the fundamental roles of actin cytoskeleton and ABPs on mitochondrial trafficking, dynamics, biogenesis, metabolism and apoptosis, with a particular focus on Gelsolin involvement in mitochondrial disorders. The functional interplay between the actin cytoskeleton, ABPs and mitochondrial membranes for the regulation of cellular homeostasis thus emerges as a new exciting field for future research and therapeutic approaches.
Collapse
Affiliation(s)
- María Illescas
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Ana Peñas
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Joaquín Arenas
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Miguel A Martín
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Cristina Ugalde
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| |
Collapse
|
6
|
Lee HJ, Kim MJ, Kim YS, Choi MY, Cho GJ, Choi WS. UHRF1 silences gelsolin to inhibit cell death in early stage cervical cancer. Biochem Biophys Res Commun 2020; 526:1061-1068. [PMID: 32312517 DOI: 10.1016/j.bbrc.2020.03.185] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 03/31/2020] [Indexed: 12/15/2022]
Abstract
Persistent infection with high-risk strains of human papillomavirus (HPV) is the primary cause of cervical cancer, the fourth most common cancer among women worldwide. Two oncoproteins encoded by the HPV genome, E6 and E7, are required for epigenetic modifications that promote cervical cancer development. We found that knockdown of HPV E6/E7 by siRNA reduced the levels of ubiquitin-like containing PHD and RING finger domain 1 (UHRF1) but increased the levels of gelsolin (GSN) in early stage cervical cancer cells. In addition, we found that UHRF1 levels were increased and GSN levels were decreased in early stage cervical cancer compared with those in normal cervical tissues, as shown by Western blot analysis, immunohistochemistry, and analysis of the Oncomine database. Moreover, knockdown of UHRF1 resulted in increased cell death in cervical cancer cell lines. Treatment of E6/E7-transformed HaCaT (HEK001) cells and HeLa cells with the DNA-hypomethylating agent 5-aza-2'-deoxycytidine and the histone deacetylase inhibitor Trichostatin A increased GSN expression levels. UHRF1 knockdown in HEK001 cells by siRNA or the UHRF1 antagonist thymoquinone increased GSN levels, induced cell cycle arrest and apoptosis, and increased the levels of p27 and cleaved PARP. Those results indicate that upregulation of UHRF1 by HPV E6/E7 causes GSN silencing and a reduction of cell death in early stage cervical cancer, suggesting that GSN might be a useful therapeutic target in early stage cervical cancer.
Collapse
Affiliation(s)
- Han Ju Lee
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Min Jun Kim
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Yoon Sook Kim
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Mee Young Choi
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Gyeong Jae Cho
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Wan Sung Choi
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea.
| |
Collapse
|
7
|
Chen Z, Li K, Yin X, Li H, Li Y, Zhang Q, Wang H, Qiu Y. Lower Expression of Gelsolin in Colon Cancer and Its Diagnostic Value in Colon Cancer Patients. J Cancer 2019; 10:1288-1296. [PMID: 30854138 PMCID: PMC6400693 DOI: 10.7150/jca.28529] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 01/04/2019] [Indexed: 12/26/2022] Open
Abstract
Colon cancer is one of the most common malignancies causing the majority of cancer-related deaths. Gelsolin (GSN) has been found to be dysregulated in various cancers. However, the secreted GSN in colon cancer remains largely unknown. In the present study, we explored the expression profile of GSN in colon cancer tissues and the diagnostic value of serum GSN in colon cancer. In addition, the effects of secreted GSN in colon cancer cells were studied. We thus found that immunoreactive GSN levels were significantly lower in colon cancer tissues than those in non-tumor colon tissues. Functional studies demonstrated that secreted GSN could restrain cell invasion and migration in vitro. Mechanistically, dose dependent recombinant GSN down-regulated the expression of MMP2 and MMP9, which might restrain the process of cell invasion and migration. Furthermore, serum levels of GSN were significantly lower in colon cancer patients than those in healthy volunteers, and ROC curves showed serum level of GSN had a better diagnostic value for colon cancer (AUC=0.932) than the traditional tumor biomarker Carcinoembryonic Antigen (CEA) or Carbohydrate Antigen 19-9 (CA199). In conclusion, our results suggest that the secreted GSN restrains the invasion and migration of colon cancer cells. Meanwhile, the serum GSN may be a new biomarker for the diagnosis of colon cancer.
Collapse
Affiliation(s)
- Zhuoyu Chen
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Clinical Laboratory, Beijing University of Chinese Medicine Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Kaifei Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaofeng Yin
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Haixia Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yao Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiong Zhang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Haifang Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yurong Qiu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Guangdong Key Laboratory of Biochip Technology, Southern Medical University, Guangzhou, Guangdong, China.,Huayin Medical Laboratory Center Co., Ltd., Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Soekmadji C, Rockstroh A, Ramm GA, Nelson CC, Russell PJ. Extracellular Vesicles in the Adaptive Process of Prostate Cancer during Inhibition of Androgen Receptor Signaling by Enzalutamide. Proteomics 2018; 17. [PMID: 29105980 DOI: 10.1002/pmic.201600427] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 09/11/2017] [Indexed: 01/05/2023]
Abstract
Current treatments for advanced prostate cancer focus on inhibition of the androgen receptor (AR) by androgen deprivation therapy (ADT). However, complex interactions mediated by tumor suppressors, oncogenes, aberrations of AR expression, or de novo androgen production have been shown to induce the adaptive response of prostate cancer, leading to the development of castration resistant prostate cancer. In this study, we report the effects of AR antagonist, enzalutamide on the protein contents of extracellular vesicles (EVs). EVs mediate cell-to-cell communication and increasing evidence shows the role of EVs in promoting cancer survival and metastasis. We found that treatment with enzalutamide alters the secretion of EVs, one of which is a plasma membrane calcium pump, ATP2B1/PMCA ATPase, as an AR-regulated EV protein. We highlight the networks of interactions between AR, Ca2+ , and ATP2B1, where the extracellular proteins thrombospondin-1, gelsolin, and integrinß1 were previously reported as regulators for cancer progression and metastasis, indicating the potential role of EV-derived proteins in mediating calcium homoeostasis under AR inhibition by enzalutamide. Our data further highlight the cross-talk between AR signaling and EV pathways in mediating resistance toward ADT.
Collapse
Affiliation(s)
- Carolina Soekmadji
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Anja Rockstroh
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology (QUT),, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland, Australia
| | - Grant A Ramm
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Colleen C Nelson
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology (QUT),, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland, Australia
| | - Pamela J Russell
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology (QUT),, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
9
|
Borlak J, Länger F, Chatterji B. Serum proteome mapping of EGF transgenic mice reveal mechanistic biomarkers of lung cancer precursor lesions with clinical significance for human adenocarcinomas. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3122-3144. [PMID: 29960043 DOI: 10.1016/j.bbadis.2018.06.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/12/2018] [Accepted: 06/25/2018] [Indexed: 12/18/2022]
Abstract
Atypical adenomatous hyperplasia (AAH) of the lung is a pre-invasive lesion (PL) with high risk of progression to lung cancer (LC). However, the pathways involved are uncertain. We searched for novel mechanistic biomarkers of AAH in an EGF transgenic disease model of lung cancer. Disease regulated proteins were validated by Western immunoblotting and immunohistochemistry (IHC) of control and morphologically altered respiratory epithelium. Translational work involved clinical resection material. Collectively, 68 unique serum proteins were identified by 2DE-MALDI-TOF mass spectrometry and 13 reached statistical significance (p < 0.05). EGF, amphiregulin and the EGFR endosomal sorting protein VPS28 were induced up to 5-fold while IHC confirmed strong induction of these proteins. Furthermore, ApoA1, α-2-macroglobulin, and vitamin-D binding protein were nearly 6- and 2-fold upregulated in AAH; however, ApoA1 was oppositely regulated in LC to evidence disease stage dependent regulation of this tumour suppressor. Conversely, plasminogen and transthyretin were highly significantly repressed by 3- and 20-fold. IHC confirmed induced ApoA1, Fetuin-B and transthyretin expression to influence calcification, inflammation and tumour-infiltrating macrophages. Moreover, serum ApoA4, ApoH and ApoM were 2-, 2- and 6-fold repressed; however tissue ApoM and sphingosine-1-phosphate receptor expression was markedly induced to suggest a critical role of sphingosine-1-phosphate signalling in PL and malignant transformation. Finally, a comparison of three different LC models revealed common and unique serum biomarkers mechanistically linked to EGFR, cMyc and cRaf signalling. Their validation by IHC on clinical resection material established relevance for distinct human lung pathologies. In conclusion, we identified mechanistic biomarker candidates recommended for in-depth clinical evaluation.
Collapse
Affiliation(s)
- Jürgen Borlak
- Hannover Medical School, Centre for Pharmacology and Toxicology, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Florian Länger
- Hannover Medical School, Institute of Pathology, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Bijon Chatterji
- Hannover Medical School, Centre for Pharmacology and Toxicology, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
10
|
Yuan X, Wang W, Li J, Zheng P, Dong P, Chen L, Zhou Y, Xie G, Xu D, Liu Y, Shen L. Gelsolin suppresses gastric cancer metastasis through inhibition of PKR-p38 signaling. Oncotarget 2018; 7:53459-53470. [PMID: 27419625 PMCID: PMC5288199 DOI: 10.18632/oncotarget.10557] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/05/2016] [Indexed: 01/22/2023] Open
Abstract
The biological function of gelsolin in gastric cancer and its mechanism remained undefined. Here, we demonstrated that gelsolin was down-regulated in human gastric cancer tissues, and lower tumorous gelsolin significantly correlated with gastric cancer metastasis. Functionally, gelsolin suppressed the migration of gastric cancer cells in vitro and inhibited lung metastasis in vivo. In mechanism, gelsolin decreased epithelial–mesenchymal transition (EMT) inducing cytoskeleton remolding through inhibition of p38 signaling to suppress the migration of gastric cancer cell. Moreover, gelsolin bound to and decreased the phosphorylation of PKR, and then inhibited p38 signaling pathway. Finally, similar to the gastric cancer cell lines, PKR-p38 signaling pathway proteins tend to be activated and correlated with low expression of gelsolin in clinical gastric cancer tissues. Altogether, these results highlight the importance of gelsolin in suppression of gastric cancer metastasis through inhibition of PKR-p38 signaling pathway.
Collapse
Affiliation(s)
- Xiangliang Yuan
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Weiwei Wang
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Junhua Li
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Peiming Zheng
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Ping Dong
- Department of Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Lei Chen
- Department of Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yunlan Zhou
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Guohua Xie
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Dakang Xu
- MIMR-PHI Institute of Medical Research, Monash University, Clayton, Victoria 3168, Australia.,Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou, Zhejiang 311121, China
| | - Yingbin Liu
- Department of Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Lisong Shen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
11
|
Shi M, Zhou L, Zhao L, Shang M, He T, Tang Z, Sun H, Ren P, Lin Z, Chen T, Yu J, Xu J, Yu X, Huang Y. Csseverin inhibits apoptosis through mitochondria-mediated pathways triggered by Ca2 + dyshomeostasis in hepatocarcinoma PLC cells. PLoS Negl Trop Dis 2017; 11:e0006074. [PMID: 29125839 PMCID: PMC5705155 DOI: 10.1371/journal.pntd.0006074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/28/2017] [Accepted: 10/26/2017] [Indexed: 01/08/2023] Open
Abstract
Background Numerous experimental and epidemiological studies have demonstrated a link between Clonorchis sinensis (C. sinensis) infestation and cholangiocarcinoma (CCA) as well as hepatocellular carcinoma (HCC). The underlying molecular mechanism involved in the malignancy of CCA and HCC has not yet been addressed. Csseverin, a component of the excretory/secretory products of C. sinensis (CsESPs), was confirmed to cause obvious apoptotic inhibition in the human HCC cell line PLC. However, the antiapoptotic mechanism is unclear. In the present study, we investigated the cellular features of the antiapoptotic mechanism upon transfection of the Csseverin gene. Methods In the present study, we evaluated the effects of Csseverin gene overexpression on the apoptosis of PLC cells using an Annexin PE/7-AAD assay. Western blotting was applied to quantify the activation of caspase-3 and caspase-9, the mitochondrial translocation of Bax and the release of Cyt c upon Csseverin overexpression in PLC cells. Laser scanning confocal microscopy was used to analyze the changes of intracellular calcium. Fluorescence assay and immunofluorescence assays were performed to observe the changes of the mitochondrial permeability transition pore (MPTP). Results The overexpression of Csseverin in PLC cells showed apoptosis resistance after the induction of apoptosis. Additionally, the activation of caspase-3 and caspase-9 was specifically weakened in Csseverin overexpression PLC cells. The overexpression of Csseverin reduced the increase in intracellular free Ca2+, thereby inhibiting MPTP opening in PLC cells. Moreover, Bax mitochondrial translocation and the subsequent release of Cyt c were downregulated in apoptotic Csseverin overexpression PLC cells. Conclusions The present findings suggest that Csseverin, a component of CsESPs, confers protection from human HCC cell apoptosis via the inactivation of membranous Ca2+ channels. Csseverin might be involved in the process of HCC through C. sinensis infestation in affected patients. Multiple studies have contributed to the association between Clonorchis sinensis (C. sinensis) infestation and cholangiocarcinoma (CCA) as well as hepatocellular carcinoma (HCC) in past years. However, studies on the underlying pathogenic mechanisms of C. sinensis lag behind those of other parasitic diseases. The excretory/secretory products of C. sinensis (CsESPs) are pathogenic, as these products promote cell proliferation, suppress cell apoptosis and stimulate inflammation. Csseverin, a component of CsESPs, inhibited the apoptosis of the human HCC cell line PLC in our previous study. The present study illustrated that Csseverin conferred human HCC cells protection from apoptosis via an intrinsic pathway (mitochondrial-mediated) triggered by the inactivation of membranous Ca2+ channels.
Collapse
Affiliation(s)
- Mengchen Shi
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lina Zhou
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Lu Zhao
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Mei Shang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Tongtong He
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Zeli Tang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Hengchang Sun
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Pengli Ren
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Zhipeng Lin
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Tingjin Chen
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Jinyun Yu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Jin Xu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Xinbing Yu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
- * E-mail: (XY); (YH)
| | - Yan Huang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
- * E-mail: (XY); (YH)
| |
Collapse
|
12
|
Serio G, Pezzuto F, Marzullo A, Scattone A, Cavone D, Punzi A, Fortarezza F, Gentile M, Buonadonna AL, Barbareschi M, Vimercati L. Peritoneal Mesothelioma with Residential Asbestos Exposure. Report of a Case with Long Survival (Seventeen Years) Analyzed by Cgh-Array. Int J Mol Sci 2017; 18:E1818. [PMID: 28829357 PMCID: PMC5578204 DOI: 10.3390/ijms18081818] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/17/2017] [Accepted: 08/18/2017] [Indexed: 12/18/2022] Open
Abstract
Malignant mesothelioma is a rare and aggressive tumor with limited therapeutic options. We report a case of a malignant peritoneal mesothelioma (MPM) epithelioid type, with environmental asbestos exposure, in a 36-year-old man, with a long survival (17 years). The patient received standard treatment which included cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC). METHODS AND RESULTS Molecular analysis with comparative genomic hybridization (CGH)-array was performed on paraffin-embedded tumoral samples. Multiple chromosomal imbalances were detected. The gains were prevalent. Losses at 1q21, 2q11.1→q13, 8p23.1, 9p12→p11, 9q21.33→q33.1, 9q12→q21.33, and 17p12→p11.2 are observed. Chromosome band 3p21 (BAP1), 9p21 (CDKN2A) and 22q12 (NF2) are not affected. Conclusions: the defects observed in this case are uncommon in malignant peritoneal mesothelioma. Some chromosomal aberrations that appear to be random here, might actually be relevant events explaining the response to therapy, the long survival and, finally, may be considered useful prognostic factors in peritoneal malignant mesothelioma (PMM).
Collapse
Affiliation(s)
- Gabriella Serio
- Department of Emergency and Organ Transplantation, Division of Pathology, Medical School, University of Bari, 11 G. Cesare Square, 70124 Bari, Italy.
| | - Federica Pezzuto
- Department of Emergency and Organ Transplantation, Division of Pathology, Medical School, University of Bari, 11 G. Cesare Square, 70124 Bari, Italy.
| | - Andrea Marzullo
- Department of Emergency and Organ Transplantation, Division of Pathology, Medical School, University of Bari, 11 G. Cesare Square, 70124 Bari, Italy.
| | - Anna Scattone
- Division of Pathology, IRCCS, National Cancer Institute "Giovanni Paolo II", 70124 Bari, Italy.
| | - Domenica Cavone
- Department of Interdisciplinary Medicine, Occupational Health Division, Medical School, University of Bari, 70124 Bari, Italy.
| | - Alessandra Punzi
- Department of Emergency and Organ Transplantation, Division of Pathology, Medical School, University of Bari, 11 G. Cesare Square, 70124 Bari, Italy.
| | - Francesco Fortarezza
- Department of Emergency and Organ Transplantation, Division of Pathology, Medical School, University of Bari, 11 G. Cesare Square, 70124 Bari, Italy.
| | - Mattia Gentile
- Division of Medical Genetics "Di Venere Hospital", 70124 Bari, Italy.
| | | | | | - Luigi Vimercati
- Department of Interdisciplinary Medicine, Occupational Health Division, Medical School, University of Bari, 70124 Bari, Italy.
| |
Collapse
|
13
|
Zhang L, Han C, Ye F, He Y, Jin Y, Wang T, Wu Y, Jiang Y, Zhang F, Jin X. Plasma Gelsolin Induced Glomerular Fibrosis via the TGF-β1/Smads Signal Transduction Pathway in IgA Nephropathy. Int J Mol Sci 2017; 18:ijms18020390. [PMID: 28208683 PMCID: PMC5343925 DOI: 10.3390/ijms18020390] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 01/29/2017] [Accepted: 02/08/2017] [Indexed: 11/16/2022] Open
Abstract
Glomerular fibrosis has been shown to be closely related to the progression and prognosis of IgA nephropathy (IgAN). However, mechanism underlying IgAN glomerular fibrosis remains unclear. Recently, our study showed that plasma gelsolin (pGSN) was decreased in the serum of an IgAN mouse model and that pGSN deposition was found in the glomeruli. Another cytokine, TGF-β1, which is closely related to glomerular fibrosis, was also found to be highly expressed in the glomeruli. In the present study, we report that pGSN induces glomerular fibrosis through the TGF-β1/Smads signal transduction pathway. This is supported by the following findings: human mesangial cells (HMCs) show remarkable morphological changes and proliferation in response to co-stimulation with pGSN and polymeric IgA1 (pIgA1) from IgAN patients compared to other controls. Moreover, ELISA assays showed that more TGF-β1 secretion was found in HMCs supernatants in the co-stimulation group. Further experiments showed increased TGF-β1, Smad3, p-Smad2/3, Smad4, and collagen 1 and decreased Smad7 expression in the co-stimulation group. Our present study implied that the synergistic effect of pGSN and pIgA induced glomerular fibrosis via the TGF-β1/Smads signal transduction pathway. This might be a potential mechanism for the glomerular fibrosis observed in IgAN patients.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| | - Changsong Han
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| | - Fei Ye
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| | - Yan He
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| | - Yinji Jin
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| | - Tianzhen Wang
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| | - Yiqi Wu
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| | - Yang Jiang
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| | - Fengmin Zhang
- Department of Microbiology, Harbin Medical University, Harbin 150081, China.
| | - Xiaoming Jin
- Department of Pathology, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
14
|
Deng R, Hao J, Han W, Ni Y, Huang X, Hu Q. Gelsolin regulates proliferation, apoptosis, migration and invasion in human oral carcinoma cells. Oncol Lett 2015; 9:2129-2134. [PMID: 26137026 DOI: 10.3892/ol.2015.3002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 11/28/2014] [Indexed: 12/14/2022] Open
Abstract
Gelsolin (GSN) is one of the most abundant actin-binding proteins, and is involved in several pathological processes, including Alzheimer's disease, cardiac injury and cancer. The aim of the present study was to assess the effect of GSN on the growth and motility of oral squamous cell carcinoma Tca8113 cells. The overexpression vector pcDNA3.1-GSN was transfected into Tca8113 cells and the stable GSN overexpression cell line was identified based on G418 antibiotic selection. The effect of GSN overexpression on the proliferation, apoptosis, migration and invasion of Tca8113 cells was examined using a cell counting kit-8 assay, flow cytometry and Transwell assays. The results revealed that GSN overexpression significantly promoted the cell proliferation and apoptosis of Tca8113 cells. In addition, Transwell assays demonstrated that the migration and invasion abilities of Tca8113 cells were enhanced by GSN overexpression. Therefore, the upregulation of GSN promotes cell growth and motility, indicating that it may perform a vital function in the progression of human oral cancers.
Collapse
Affiliation(s)
- Runzhi Deng
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Medical School, Nanjing University, Nanjing, Jiangsu, P.R. China
| | - Jing Hao
- Central Laboratory of Nanjing Stomatological Hospital, Nanjing, Jiangsu, P.R. China
| | - Wei Han
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Medical School, Nanjing University, Nanjing, Jiangsu, P.R. China ; Central Laboratory of Nanjing Stomatological Hospital, Nanjing, Jiangsu, P.R. China
| | - Yanhong Ni
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Medical School, Nanjing University, Nanjing, Jiangsu, P.R. China ; Central Laboratory of Nanjing Stomatological Hospital, Nanjing, Jiangsu, P.R. China
| | - Xiaofeng Huang
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Medical School, Nanjing University, Nanjing, Jiangsu, P.R. China
| | - Qingang Hu
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Medical School, Nanjing University, Nanjing, Jiangsu, P.R. China
| |
Collapse
|
15
|
Kanda M, Nomoto S, Oya H, Hashimoto R, Takami H, Shimizu D, Sonohara F, Kobayashi D, Tanaka C, Yamada S, Fujii T, Nakayama G, Sugimoto H, Koike M, Murotani K, Fujiwara M, Kodera Y. Decreased expression of prenyl diphosphate synthase subunit 2 correlates with reduced survival of patients with gastric cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:88. [PMID: 25330808 PMCID: PMC4209044 DOI: 10.1186/s13046-014-0088-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 10/03/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND Identification of novel molecular biomarkers will improve the management of patients with gastric cancer (GC). Prenyl diphosphate synthase subunit 2 (PDSS2) is required for coenzyme Q10 biosynthesis and acts as a tumor suppressor; however, the role and regulatory mechanisms of PDSS2 in GC are not understood. The aim of this study was to determine expression status and regulatory mechanisms of PDSS2 in GC. METHODS Associations between expression and methylation of PDSS2 were evaluated using GC cell lines. The clinical significance of PDSS2 expression was evaluated using 238 pairs of surgically resected gastric tissues with subgroup analysis based on GC subtypes. RESULTS The expression of PDSS2 mRNA was decreased in 73% of GC cell lines compared with the control non-cancerous cell. The PDSS2 promoter was hypermethylated in cells with decreased PDSS2 expression, and treating these cells with a methylation inhibitor reactivated PDSS2 expression. GC tissues expressed significantly lower mean levels of PDSS2 mRNA compared with adjacent normal tissues (P <0.001). The expression pattern of PDSS2 protein was consistent with that of its mRNA. The decrease of PDSS2 mRNA expression in GC tissues (less than half the level of expression detected in the corresponding normal adjacent tissues) correlated significantly with elevated levels of carbohydrate antigen 19-9 (P = 0.015), lymph node metastasis (P = 0.022), and shorter recurrence-free survival after curative resection (P = 0.022). Further, multivariate analysis identified PDSS2 mRNA expression as an independent prognostic factor (hazard ratio 1.95, 95% confidence interval 1.22-3.09, P = 0.005), and its expression pattern and prognostic significance were similar among three GC subtypes. CONCLUSIONS PDSS2 encodes a putative tumor suppressor, and we show here that its expression was regulated by hypermethylation of its promoter in GC cells. Inhibition of PDSS2 mRNA expression may serve as a novel biomarker of all types of GC.
Collapse
Affiliation(s)
- Mitsuro Kanda
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Shuji Nomoto
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Hisaharu Oya
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Ryoji Hashimoto
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Hideki Takami
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Dai Shimizu
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Fuminori Sonohara
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Daisuke Kobayashi
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Chie Tanaka
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Suguru Yamada
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Tsutomu Fujii
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Goro Nakayama
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Hiroyuki Sugimoto
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Masahiko Koike
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Kenta Murotani
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan.
| | - Michitaka Fujiwara
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| |
Collapse
|
16
|
Kanda M, Nomoto S, Oya H, Hashimoto R, Takami H, Shimizu D, Sonohara F, Kobayashi D, Tanaka C, Yamada S, Fujii T, Nakayama G, Sugimoto H, Koike M, Murotani K, Fujiwara M, Kodera Y. Decreased expression of prenyl diphosphate synthase subunit 2 correlates with reduced survival of patients with gastric cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014. [PMID: 25330808 DOI: 10.1186/preaccept-8549609481376418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Identification of novel molecular biomarkers will improve the management of patients with gastric cancer (GC). Prenyl diphosphate synthase subunit 2 (PDSS2) is required for coenzyme Q10 biosynthesis and acts as a tumor suppressor; however, the role and regulatory mechanisms of PDSS2 in GC are not understood. The aim of this study was to determine expression status and regulatory mechanisms of PDSS2 in GC. METHODS Associations between expression and methylation of PDSS2 were evaluated using GC cell lines. The clinical significance of PDSS2 expression was evaluated using 238 pairs of surgically resected gastric tissues with subgroup analysis based on GC subtypes. RESULTS The expression of PDSS2 mRNA was decreased in 73% of GC cell lines compared with the control non-cancerous cell. The PDSS2 promoter was hypermethylated in cells with decreased PDSS2 expression, and treating these cells with a methylation inhibitor reactivated PDSS2 expression. GC tissues expressed significantly lower mean levels of PDSS2 mRNA compared with adjacent normal tissues (P <0.001). The expression pattern of PDSS2 protein was consistent with that of its mRNA. The decrease of PDSS2 mRNA expression in GC tissues (less than half the level of expression detected in the corresponding normal adjacent tissues) correlated significantly with elevated levels of carbohydrate antigen 19-9 (P = 0.015), lymph node metastasis (P = 0.022), and shorter recurrence-free survival after curative resection (P = 0.022). Further, multivariate analysis identified PDSS2 mRNA expression as an independent prognostic factor (hazard ratio 1.95, 95% confidence interval 1.22-3.09, P = 0.005), and its expression pattern and prognostic significance were similar among three GC subtypes. CONCLUSIONS PDSS2 encodes a putative tumor suppressor, and we show here that its expression was regulated by hypermethylation of its promoter in GC cells. Inhibition of PDSS2 mRNA expression may serve as a novel biomarker of all types of GC.
Collapse
Affiliation(s)
- Mitsuro Kanda
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Shuji Nomoto
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Hisaharu Oya
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Ryoji Hashimoto
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Hideki Takami
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Dai Shimizu
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Fuminori Sonohara
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Daisuke Kobayashi
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Chie Tanaka
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Suguru Yamada
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Tsutomu Fujii
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Goro Nakayama
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Hiroyuki Sugimoto
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Masahiko Koike
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Kenta Murotani
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan.
| | - Michitaka Fujiwara
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| |
Collapse
|
17
|
Hohenester S, Vennegeerts T, Wagner M, Wimmer R, Drolle H, Rieger C, Denk GU, Rust C, Fiegl M. Physiological hypoxia prevents bile salt-induced apoptosis in human and rat hepatocytes. Liver Int 2014; 34:1224-31. [PMID: 24164780 DOI: 10.1111/liv.12368] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/20/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Hydrophobic bile salts such as glycochenodeoxycholate (GCDC) accumulate in cholestatic liver disease and induce hepatocellular apoptosis, promoting profibrotic signalling. The tissue microenvironment is an integral player in cellular pathophysiology, but it is not routinely incorporated into laboratory studies. Tissue oxygen partial pressure (pO₂) may be an underestimated component of the microenvironment: in the liver, a pO₂ of 30-45 mmHg (approximately 6% O₂) is physiological, because of predominant portal blood supply. It was the aim of this project to investigate the impact of physiological hypoxia (i.e. 6% O₂) on hepatocellular function, namely, bile salt-induced apoptosis. METHODS Human hepatoma cells (HepG2-Ntcp) and primary rat hepatocytes were cultured at standard laboratory (hyperoxic) conditions (21% O₂) and at physiological hypoxia (6% O₂) in parallel for 1-8 days to study hepatocellular apoptosis and activation of signalling pathways. Standard laboratory analyses were applied for bile salt uptake, caspase-3/-7 activity, western blotting and gene-array analysis. RESULTS Culturing at physiological hypoxia protected both human and rat hepatocytes against GCDC-induced apoptosis: caspase-3/-7 activation was diminished by 3.1 ± 0.5-fold in human HepG2-Ntcp and completely abolished in primary rat hepatocytes. Bile salt uptake was unaffected. Induction of hypoxia-inducible factor-1α indicated adaption to physiological hypoxia. The MEK/ERK cascade was activated and anti-apoptotic mediators were induced: N-Myc down-regulated gene, gelsolin and carbonic anhydrase IX were upregulated 12.4-, 6.5- and 5.2-fold respectively. CONCLUSIONS We conclude from these data that (i) physiological hypoxia protects hepatocytes from bile salt-induced apoptosis, (ii) tissue pO₂ is a crucial, underestimated component of the microenvironment and should (iii) be considered when studying hepatocellular physiology in vitro.
Collapse
Affiliation(s)
- Simon Hohenester
- Department of Medicine II, University of Munich, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Giantin M, Granato A, Baratto C, Marconato L, Vascellari M, Morello EM, Vercelli A, Mutinelli F, Dacasto M. Global gene expression analysis of canine cutaneous mast cell tumor: could molecular profiling be useful for subtype classification and prognostication? PLoS One 2014; 9:e95481. [PMID: 24748173 PMCID: PMC3991658 DOI: 10.1371/journal.pone.0095481] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 03/27/2014] [Indexed: 02/06/2023] Open
Abstract
Prognosis and therapeutic management of dogs with cutaneous mast cell tumors (MCTs) depend on clinical stage and histological grade. However, the prognostic value of this latter is still questionable. In the present study, MCT transcriptome was analyzed to identify a set of candidate genes potentially useful for predicting the biological behavior of MCTs. Fifty-one canine MCT biopsies were analyzed. Isolated and purified total RNAs were individually hybridized to the Agilent Canine V2 4x44k DNA microarray. The comparison of reference differentiated and undifferentiated MCT transcriptome revealed a total of 597 differentially expressed genes (147 down-regulated and 450 up-regulated). The functional analysis of this set of genes provided evidence that they were mainly involved in cell cycle, DNA replication, p53 signaling pathway, nucleotide excision repair and pyrimidine metabolism. Class prediction analysis identified 13 transcripts providing the greatest accuracy of class prediction and divided samples into two categories (differentiated and undifferentiated), harboring a different prognosis. The Principal Component Analysis of all samples, made by using the selected 13 markers, confirmed MCT classification. The first three components accounted for 99.924% of the total variance. This molecular classification significantly correlated with survival time (p = 0.0026). Furthermore, among all marker genes, a significant association was found between mRNA expression and MCT-related mortality for FOXM1, GSN, FEN1 and KPNA2 (p<0.05). Finally, marker genes mRNA expression was evaluated in a cohort of 22 independent samples. Data obtained enabled to identify MCT cases with different prognosis. Overall, the molecular characterization of canine MCT transcriptome allowed the identification of a set of 13 transcripts that clearly separated differentiated from undifferentiated MCTs, thus predicting outcome regardless of the histological grade. These results may have clinical relevance and warrant future validation in a prospective study.
Collapse
Affiliation(s)
- Mery Giantin
- Dipartimento di Biomedicina Comparata e Alimentazione, Università di Padova, Legnaro (Padova), Italy
- * E-mail:
| | - Anna Granato
- Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (Padova), Italy
| | - Chiara Baratto
- Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (Padova), Italy
| | - Laura Marconato
- Centro Oncologico Veterinario, Sasso Marconi, Bologna, Italy
| | - Marta Vascellari
- Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (Padova), Italy
| | - Emanuela M. Morello
- Dipartimento di Scienze Veterinarie, Università di Torino, Grugliasco (Torino), Italy
| | | | - Franco Mutinelli
- Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (Padova), Italy
| | - Mauro Dacasto
- Dipartimento di Biomedicina Comparata e Alimentazione, Università di Padova, Legnaro (Padova), Italy
| |
Collapse
|
19
|
Chen X, Li S, He L, Wang X, Liang P, Chen W, Bian M, Ren M, Lin J, Liang C, Xu J, Wu Z, Li X, Huang Y, Yu X. Molecular characterization of severin from Clonorchis sinensis excretory/secretory products and its potential anti-apoptotic role in hepatocarcinoma PLC cells. PLoS Negl Trop Dis 2013; 7:e2606. [PMID: 24367717 PMCID: PMC3868641 DOI: 10.1371/journal.pntd.0002606] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 10/28/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Clonorchiasis, caused by the infection of Clonorchis sinensis (C. sinensis), is a kind of neglected tropical disease, but it is highly related to cholangiocarcinoma and hepatocellular carcinoma (HCC). It has been well known that the excretory/secretory products of C. sinensis (CsESPs) play key roles in clonorchiasis associated carcinoma. From genome and transcriptome of C. sinensis, we identified one component of CsESPs, severin (Csseverin), which had three putative gelsolin domains. Its homologues are supposed to play a vital role in apoptosis resistance of tumour cell. METHODOLOGY/PRINCIPAL FINDINGS There was significant similarity in tertiary structures between human gelsolin and Csseverin by bioinformatics analysis. We identified that Csseverin expressed at life stage of adult worm, metacercaria and egg by the method of quantitative real-time PCR and western blotting. Csseverin distributed in vitellarium and intrauterine eggs of adult worm and tegument of metacercaria by immunofluorence assay. We obtained recombinant Csseverin (rCsseverin) and confirmed that rCsseverin could bind with calciumion in circular dichroism spectrum analysis. It was demonstrated that rCsseverin was of the capability of actin binding by gel overlay assay and immunocytochemistry. Both Annexin V/PI assay and mitochondrial membrane potential assay of human hepatocarcinoma cell line PLC showed apoptosis resistance after incubation with different concentrations of rCsseverin. Morphological analysis, apoptosis-associated changes of mitochondrial membrane potential and Annexin V/PI apoptosis assay showed that co-incubation of PLC cells with rCsseverin in vitro led to an inhibition of apoptosis induced by serum-starved for 24 h. CONCLUSIONS/SIGNIFICANCE Collectively, the molecular properties of Csseverin, a molecule of CsESPs, were characterized in our study. rCsseverin could cause obvious apoptotic inhibition in human HCC cell line. Csseverin might exacerbate the process of HCC patients combined with C. sinensis infection.
Collapse
Affiliation(s)
- Xueqing Chen
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People′s Republic of China
- Key Laboratory of Tropical Diseases Control at Sun Yat-sen University, Ministry of Education, Guangzhou, People′s Republic of China
| | - Shan Li
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People′s Republic of China
- Key Laboratory of Tropical Diseases Control at Sun Yat-sen University, Ministry of Education, Guangzhou, People′s Republic of China
| | - Lei He
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People′s Republic of China
- Key Laboratory of Tropical Diseases Control at Sun Yat-sen University, Ministry of Education, Guangzhou, People′s Republic of China
| | - Xiaoyun Wang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People′s Republic of China
- Key Laboratory of Tropical Diseases Control at Sun Yat-sen University, Ministry of Education, Guangzhou, People′s Republic of China
| | - Pei Liang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People′s Republic of China
- Key Laboratory of Tropical Diseases Control at Sun Yat-sen University, Ministry of Education, Guangzhou, People′s Republic of China
| | - Wenjun Chen
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People′s Republic of China
- Key Laboratory of Tropical Diseases Control at Sun Yat-sen University, Ministry of Education, Guangzhou, People′s Republic of China
| | - Meng Bian
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People′s Republic of China
- Key Laboratory of Tropical Diseases Control at Sun Yat-sen University, Ministry of Education, Guangzhou, People′s Republic of China
| | - Mengyu Ren
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People′s Republic of China
- Key Laboratory of Tropical Diseases Control at Sun Yat-sen University, Ministry of Education, Guangzhou, People′s Republic of China
| | - Jinsi Lin
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People′s Republic of China
- Key Laboratory of Tropical Diseases Control at Sun Yat-sen University, Ministry of Education, Guangzhou, People′s Republic of China
| | - Chi Liang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People′s Republic of China
- Key Laboratory of Tropical Diseases Control at Sun Yat-sen University, Ministry of Education, Guangzhou, People′s Republic of China
| | - Jin Xu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People′s Republic of China
- Key Laboratory of Tropical Diseases Control at Sun Yat-sen University, Ministry of Education, Guangzhou, People′s Republic of China
| | - Zhongdao Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People′s Republic of China
- Key Laboratory of Tropical Diseases Control at Sun Yat-sen University, Ministry of Education, Guangzhou, People′s Republic of China
| | - Xuerong Li
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People′s Republic of China
- Key Laboratory of Tropical Diseases Control at Sun Yat-sen University, Ministry of Education, Guangzhou, People′s Republic of China
| | - Yan Huang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People′s Republic of China
- Key Laboratory of Tropical Diseases Control at Sun Yat-sen University, Ministry of Education, Guangzhou, People′s Republic of China
| | - Xinbing Yu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People′s Republic of China
- Key Laboratory of Tropical Diseases Control at Sun Yat-sen University, Ministry of Education, Guangzhou, People′s Republic of China
| |
Collapse
|
20
|
Hu Y, Li H, Li WH, Meng HX, Fan YZ, Li WJ, Ji YT, Zhao H, Zhang L, Jin XM, Zhang FM. The value of decreased plasma gelsolin levels in patients with systemic lupus erythematosus and rheumatoid arthritis in diagnosis and disease activity evaluation. Lupus 2013; 22:1455-61. [PMID: 24122723 DOI: 10.1177/0961203313507985] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Plasma gelsolin, the extracellular gelsolin isoform, circulates in the blood of healthy individuals at a concentration of 200 ± 50 mg/l and plays important roles in the extracellular actin-scavenging system during tissue damage. Decreased plasma gelsolin levels have been observed in many inflammatory diseases. In the present study, the variation and potential clinical application of plasma gelsolin levels in patients with systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) were analysed. Plasma samples and clinical data were collected from informed and consenting participants: 47 SLE patients, 60 RA patients and 50 age- and gender-matched healthy individuals. Semiquantitative western blotting was used for measuring plasma gelsolin levels. The plasma gelsolin levels in patients with SLE and RA were significantly decreased compared with healthy controls (145.3 ± 40.4 versus 182.7 ± 38.3 mg/l and 100.8 ± 36 versus 182.7 ± 38.3 mg/l, p < 0.001), and plasma gelsolin levels were especially lower in RA than in SLE patients (100.8 ± 36 versus 145.3 ± 40.4 mg/L, p < 0.001). An analysis of the clinical data showed a significant negative correlation between plasma gelsolin levels and SLE Disease Activity Index (SLEDAI) scores (r = 0.659, p < 0.001) but no correlation between plasma gelsolin levels and RA disease activity score 28 (DAS28) (r = 0.076, p = 0.569). Different clinical characteristics were also observed in SLE and RA patients with normal and decreased plasma gelsolin levels.This study found significantly lower plasma gelsolin levels in patients with SLE and RA compared with healthy controls and documented a significant negative correlation between plasma gelsolin levels and SLEDAI, which suggested the potential clinical application of plasma gelsolin in SLE diagnosis and disease activity evaluation. The different clinical characteristics in SLE and RA patients with normal and decreased plasma gelsolin levels indicate differences in the basis of the diseases.
Collapse
Affiliation(s)
- Yl Hu
- 1Department of Microbiology, Harbin Medical University, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Marino N, Marshall JC, Collins JW, Zhou M, Qian Y, Veenstra T, Steeg PS. Nm23-h1 binds to gelsolin and inactivates its actin-severing capacity to promote tumor cell motility and metastasis. Cancer Res 2013; 73:5949-62. [PMID: 23940300 PMCID: PMC3825031 DOI: 10.1158/0008-5472.can-13-0368] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nm23-H1 has been identified as a metastasis suppressor gene, but its protein interactions have yet to be understood with any mechanistic clarity. In this study, we evaluated the proteomic spectrum of interactions made by Nm23-H1 in 4T1 murine breast cancer cells derived from tissue culture, primary mammary tumors, and pulmonary metastases. By this approach, we identified the actin-severing protein Gelsolin as binding partner for Nm23-H1, verifying their interaction by coimmunoprecipitation in 4T1 cells as well as in human MCF7, MDA-MB-231T, and MDA-MB-435 breast cancer cells. In Gelsolin-transfected cells, coexpression of Nm23-H1 abrogated the actin-severing activity of Gelsolin. Conversely, actin severing by Gelsolin was abrogated by RNA interference-mediated silencing of endogenous Nm23-H1. Tumor cell motility was negatively affected in parallel with Gelsolin activity, suggesting that Nm23-H1 binding inactivated the actin-depolymerizing function of Gelsolin to inhibit cell motility. Using indirect immunoflourescence to monitor complexes formed by Gelsolin and Nm23-H1 in living cells, we observed their colocalization in a perinuclear cytoplasmic compartment that was associated with the presence of disrupted actin stress fibers. In vivo analyses revealed that Gelsolin overexpression increased the metastasis of orthotopically implanted 4T1 or tail vein-injected MDA-MB-231T cells (P = 0.001 and 0.04, respectively), along with the proportion of mice with diffuse liver metastases, an effect ablated by coexpression of Nm23-H1. We observed no variation in proliferation among lung metastases. Our findings suggest a new actin-based mechanism that can suppress tumor metastasis.
Collapse
MESH Headings
- Actins/metabolism
- Animals
- Apoptosis
- Blotting, Western
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Female
- Fluorescent Antibody Technique
- Gelsolin/antagonists & inhibitors
- Gelsolin/genetics
- Gelsolin/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Immunoenzyme Techniques
- Immunoprecipitation
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/secondary
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/metabolism
- Mammary Neoplasms, Animal/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- NM23 Nucleoside Diphosphate Kinases/antagonists & inhibitors
- NM23 Nucleoside Diphosphate Kinases/genetics
- NM23 Nucleoside Diphosphate Kinases/metabolism
- RNA, Small Interfering/genetics
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
Collapse
Affiliation(s)
- Natascia Marino
- Women’s Cancers Section, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda MD 20892
| | - Jean-Claude Marshall
- Women’s Cancers Section, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda MD 20892
| | - Joshua W. Collins
- Women’s Cancers Section, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda MD 20892
| | - Ming Zhou
- Laboratory of Proteomics and Analytical Technologies, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Yongzhen Qian
- Women’s Cancers Section, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda MD 20892
| | - Timothy Veenstra
- Laboratory of Proteomics and Analytical Technologies, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Patricia S. Steeg
- Women’s Cancers Section, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda MD 20892
| |
Collapse
|
22
|
Marino N, Marshall JC, Collins JW, Zhou M, Qian Y, Veenstra T, Steeg PS. Nm23-h1 binds to gelsolin and inactivates its actin-severing capacity to promote tumor cell motility and metastasis. Cancer Res 2013. [PMID: 23940300 DOI: 10.1158/0008-5472.can-13-0368-009-0109-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nm23-H1 has been identified as a metastasis suppressor gene, but its protein interactions have yet to be understood with any mechanistic clarity. In this study, we evaluated the proteomic spectrum of interactions made by Nm23-H1 in 4T1 murine breast cancer cells derived from tissue culture, primary mammary tumors, and pulmonary metastases. By this approach, we identified the actin-severing protein Gelsolin as binding partner for Nm23-H1, verifying their interaction by coimmunoprecipitation in 4T1 cells as well as in human MCF7, MDA-MB-231T, and MDA-MB-435 breast cancer cells. In Gelsolin-transfected cells, coexpression of Nm23-H1 abrogated the actin-severing activity of Gelsolin. Conversely, actin severing by Gelsolin was abrogated by RNA interference-mediated silencing of endogenous Nm23-H1. Tumor cell motility was negatively affected in parallel with Gelsolin activity, suggesting that Nm23-H1 binding inactivated the actin-depolymerizing function of Gelsolin to inhibit cell motility. Using indirect immunoflourescence to monitor complexes formed by Gelsolin and Nm23-H1 in living cells, we observed their colocalization in a perinuclear cytoplasmic compartment that was associated with the presence of disrupted actin stress fibers. In vivo analyses revealed that Gelsolin overexpression increased the metastasis of orthotopically implanted 4T1 or tail vein-injected MDA-MB-231T cells (P = 0.001 and 0.04, respectively), along with the proportion of mice with diffuse liver metastases, an effect ablated by coexpression of Nm23-H1. We observed no variation in proliferation among lung metastases. Our findings suggest a new actin-based mechanism that can suppress tumor metastasis.
Collapse
MESH Headings
- Actins/metabolism
- Animals
- Apoptosis
- Blotting, Western
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Female
- Fluorescent Antibody Technique
- Gelsolin/antagonists & inhibitors
- Gelsolin/genetics
- Gelsolin/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Immunoenzyme Techniques
- Immunoprecipitation
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/secondary
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/metabolism
- Mammary Neoplasms, Animal/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- NM23 Nucleoside Diphosphate Kinases/antagonists & inhibitors
- NM23 Nucleoside Diphosphate Kinases/genetics
- NM23 Nucleoside Diphosphate Kinases/metabolism
- RNA, Small Interfering/genetics
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
Collapse
Affiliation(s)
- Natascia Marino
- Authors' Affiliations: Women's Cancers Section, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda; and Laboratory of Proteomics and Analytical Technologies, Science Applications International Corporation-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | | | | | | | | | | | | |
Collapse
|
23
|
D'Antonio M, Ciccarelli FD. Integrated analysis of recurrent properties of cancer genes to identify novel drivers. Genome Biol 2013; 14:R52. [PMID: 23718799 PMCID: PMC4054099 DOI: 10.1186/gb-2013-14-5-r52] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 05/29/2013] [Indexed: 11/10/2022] Open
Abstract
The heterogeneity of cancer genomes in terms of acquired mutations complicates the identification of genes whose modification may exert a driver role in tumorigenesis. In this study, we present a novel method that integrates expression profiles, mutation effects, and systemic properties of mutated genes to identify novel cancer drivers. We applied our method to ovarian cancer samples and were able to identify putative drivers in the majority of carcinomas without mutations in known cancer genes, thus suggesting that it can be used as a complementary approach to find rare driver mutations that cannot be detected using frequency-based approaches.
Collapse
|
24
|
Jin YL, Wang ZQ, Qu H, Wang HX, Ibrahim M, Zhang J, Huang YH, Wu J, Bai LL, Wang XY, Meng JY, Tang JW. Annexin A7 gene is an important factor in the lymphatic metastasis of tumors. Biomed Pharmacother 2013; 67:251-9. [DOI: 10.1016/j.biopha.2012.11.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 11/23/2012] [Indexed: 12/16/2022] Open
|
25
|
ICAD deficiency in human colon cancer and predisposition to colon tumorigenesis: linkage to apoptosis resistance and genomic instability. PLoS One 2013; 8:e57871. [PMID: 23451280 PMCID: PMC3579889 DOI: 10.1371/journal.pone.0057871] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 01/29/2013] [Indexed: 12/31/2022] Open
Abstract
We previously showed that DNA fragmentation factor, which comprises a caspase-3-activated DNase (CAD) and its inhibitor (ICAD), may influence the rate of cell death by generating PARP-1-activating DNA breaks. Here we tested the hypothesis that ICAD-deficient colon epithelial cells exhibiting resistance to death stimuli may accumulate additional genetic modifications, leading to a tumorigenic phenotype. We show that ICAD deficiency may be associated with colon malignancy in humans. Indeed, an examination of ICAD expression using immunohistochemistry in an array of both colon cancer and normal tissues revealed that ICAD expression levels were severely compromised in the cancerous tissues. Upon DNA damage caused by a low dose of irradiation, ICAD cells acquire a tumorigenic phenotype. Colon epithelial cells derived from ICAD mice showed a significant resistance to death induced by the colon carcinogen dimethylhydrazine in vitro and in mice. Such resistance was associated with a decrease in PARP-1 activation. In an animal model of dimethylhydrazine-induced colon tumorigenesis, ICAD−/− mice developed significantly higher numbers of tumors with markedly larger sizes than the wild-type counterparts. Interestingly, the phenotype of the ICAD−/− mice was not associated with a significant increase in the precancerous aberrant crypt foci suggesting a potential link to tumor progression rather than initiation. More importantly, ICAD deficiency was associated with severe genomic instability as assessed by array comparative genomic hybridization. Such genomic instability consisted most prominently of amplifications but with sizable deletions as compared to the wild-type counterparts affecting several cancer-related genes including RAF-1, GSN, LMO3, and Fzd6 independently of p53. Altogether, our results present a viable case for the involvement of ICAD deficiency in colon carcinogenesis and show that apoptosis and genomic instability may comprise the means by which such deficiency may contribute to the process of increasing susceptibility to carcinogen-induced tumorigenesis.
Collapse
|
26
|
Posttranslational regulation of self-renewal capacity: insights from proteome and phosphoproteome analyses of stem cell leukemia. Blood 2012; 120:e17-27. [PMID: 22802335 DOI: 10.1182/blood-2011-12-397844] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
We recently generated 2 phenotypically similar Hoxa9+Meis1 overexpressing acute myeloid leukemias that differ by their in vivo biologic behavior. The first leukemia, named FLA2, shows a high frequency of leukemia stem cells (LSCs; 1 in 1.4 cells), whereas the second, FLB1, is more typical with a frequency of LSCs in the range of 1 per several hundred cells. To gain insights into possible mechanisms that determine LSC self-renewal, we profiled and compared the abundance of nuclear and cytoplasmic proteins and phosphoproteins from these leukemias using quantitative proteomics. These analyses revealed differences in proteins associated with stem cell fate, including a hyperactive p38 MAP kinase in FLB1 and a differentially localized Polycomb group protein Ezh2, which is mostly nuclear in FLA2 and predominantly cytoplasmic in FLB1. Together, these newly documented proteomes and phosphoproteomes represent a unique resource with more than 440 differentially expressed proteins and 11 543 unique phosphopeptides, of which 80% are novel and 7% preferentially phosphorylated in the stem cell-enriched leukemia.
Collapse
|