1
|
Gopalakrishnan K, Kannan B, Pandi C, Pandi A, Ramasubramanian A, Jayaseelan VP, Arumugam P. Aberrant expression of VASP serves as a potential prognostic biomarker and therapeutic target for oral squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol 2024; 138:391-402. [PMID: 38816308 DOI: 10.1016/j.oooo.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/16/2024] [Accepted: 05/05/2024] [Indexed: 06/01/2024]
Abstract
OBJECTIVE To address the molecular markers linked to the development and progression of oral squamous cell carcinoma (OSCC), we sought to analyze the expression of vasodilator-stimulated phosphoproteins (VASP) in OSCC samples. STUDY DESIGN This study used 51 OSCC patients and The Cancer Genome Atlas-Head and Neck Squamous Cell Carcinoma (TCGA-HNSC) dataset to analyze VASP expression. The association between VASP mRNA expression and HNSCC clinicopathological features, tumor infiltration, functional roles, and gene co-expression of VASP also were evaluated. RESULTS Our study observed increased VASP mRNA expression in OSCC tumor tissues compared to normal tissues, supported by TCGA-HNSC dataset analysis. Elevated VASP levels correlated with advanced tumor stage, higher grade, nodal metastasis, and poor survival, indicating its potential as a prognostic marker. Protein analysis and immunohistochemistry confirmed these findings, and in silico analysis revealed VASP involvement in key cancer-related processes and its correlation with IL8, RAP1A expression, and tumor infiltration levels. CONCLUSIONS In conclusion, VASP emerges as a promising diagnostic and prognostic marker for OSCC within HNSCC, emphasizing the importance of exploring its regulatory mechanisms and therapeutic applications. The revealed pathways present avenues for targeted treatment in OSCC. Despite limitations, this study provides valuable insights with potential implications for improving patient outcomes.
Collapse
Affiliation(s)
- Karpakavinayakam Gopalakrishnan
- Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, TN, India
| | - Balachander Kannan
- Molecular Biology Lab, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, TN, India
| | - Chandra Pandi
- Molecular Biology Lab, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, TN, India
| | - Anitha Pandi
- Clinical Genetics Lab, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, TN, India
| | - Abilasha Ramasubramanian
- Department of Oral Pathology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, TN, India
| | - Vijayashree Priyadharsini Jayaseelan
- Clinical Genetics Lab, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, TN, India
| | - Paramasivam Arumugam
- Molecular Biology Lab, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, TN, India.
| |
Collapse
|
2
|
Tan C, Qin G, Wang QQ, Li KM, Zhou YC, Yao SK. Comprehensive serum proteomics profiles and potential protein biomarkers for the early detection of advanced adenoma and colorectal cancer. World J Gastrointest Oncol 2024; 16:2971-2987. [PMID: 39072170 PMCID: PMC11271786 DOI: 10.4251/wjgo.v16.i7.2971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/08/2024] [Accepted: 05/15/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND The majority of colorectal cancer (CRC) cases develop from precursor advanced adenoma (AA). With the development of proteomics technologies, blood protein biomarkers have potential applications in the early screening of AA and CRC in the general population. AIM To identify serum protein biomarkers for the early screening of AA and CRC. METHODS We collected 43 serum samples from 8 normal controls (NCs), 19 AA patients and 16 CRC patients at China-Japan Friendship Hospital. Quantitative proteomic analysis was performed using liquid chromatography-mass spectrometry/mass spectrometry and data independent acquisition, and differentially expressed proteins (DEPs) with P-values < 0.05 and absolute fold changes > 1.5 were screened out, followed by bioinformatics analysis. Prognosis was further analyzed based on public databases, and proteins expression in tissues were validated by immunohistochemistry. RESULTS A total of 2132 proteins and 17365 peptides were identified in the serum samples. There were 459 upregulated proteins and 118 downregulated proteins in the NC vs AA group, 289 and 180 in the NC vs CRC group, and 52 and 248 in the AA vs CRC group, respectively. Bioinformatic analysis revealed that these DEPs had different functions and participated in extensive signaling pathways. We also identified DIAPH1, VASP, RAB11B, LBP, SAR1A, TUBGCP5, and DOK3 as important proteins for the progression of AA and CRC. Furthermore, VASP (P < 0.01), LBP (P = 0.01), TUBGCP5 (P < 0.01), and DOK3 (P < 0.01) were associated with a poor prognosis. In addition, we propose that LBP and VASP may be more promising protein biomarkers for the early screening of colorectal tumors. CONCLUSION Our study elucidated the serum proteomic profiles of AA and CRC patients, and the identified proteins, such as LBP and VASP, may contribute to the early detection of AA and CRC.
Collapse
Affiliation(s)
- Chang Tan
- Graduate School, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Geng Qin
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Qian-Qian Wang
- Graduate School, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Kai-Min Li
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yuan-Chen Zhou
- Graduate School, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Shu-Kun Yao
- Graduate School, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
3
|
Fan C, Xiong F, Zhang S, Gong Z, Liao Q, Li G, Guo C, Xiong W, Huang H, Zeng Z. Role of adhesion molecules in cancer and targeted therapy. SCIENCE CHINA. LIFE SCIENCES 2024; 67:940-957. [PMID: 38212458 DOI: 10.1007/s11427-023-2417-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/17/2023] [Indexed: 01/13/2024]
Abstract
Adhesion molecules mediate cell-to-cell and cell-to-extracellular matrix interactions and transmit mechanical and chemical signals among them. Various mechanisms deregulate adhesion molecules in cancer, enabling tumor cells to proliferate without restraint, invade through tissue boundaries, escape from immune surveillance, and survive in the tumor microenvironment. Recent studies have revealed that adhesion molecules also drive angiogenesis, reshape metabolism, and are involved in stem cell self-renewal. In this review, we summarize the functions and mechanisms of adhesion molecules in cancer and the tumor microenvironment, as well as the therapeutic strategies targeting adhesion molecules. These studies have implications for furthering our understanding of adhesion molecules in cancer and providing a paradigm for exploring novel therapeutic approaches.
Collapse
Affiliation(s)
- Chunmei Fan
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, China
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Fang Xiong
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Shanshan Zhang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Zhaojian Gong
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Qianjin Liao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, China
| | - Can Guo
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, China
| | - He Huang
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, 410013, China.
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, China.
| |
Collapse
|
4
|
Gui J, Zhou H, Wan H, Yang D, Liu Q, Zhu L, Mi Y. The Role of Vasodilator-stimulated Phosphoproteins in the Development of Malignant Tumors. Curr Cancer Drug Targets 2024; 24:477-489. [PMID: 37962042 PMCID: PMC11092557 DOI: 10.2174/0115680096262439231023110106] [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: 05/15/2023] [Revised: 08/06/2023] [Accepted: 09/06/2023] [Indexed: 11/15/2023]
Abstract
Vasodilator-stimulated phosphoprotein (VASP) is an actin-binding protein that includes three structural domains: Enabled/VASP homolog1 (EVH1), EVH2, and proline-rich (PRR). VASP plays an important role in various cellular behaviors related to cytoskeletal regulation. More importantly, VASP plays a key role in the progression of several malignant tumors and is associated with malignant cell proliferation, invasion, and metastasis. Here, we have summarized current studies on the impact of VASP on the development of several malignant tumors and their mechanisms. This study provides a new theoretical basis for clinical molecular diagnosis and molecular targeted therapy.
Collapse
Affiliation(s)
- Jiandong Gui
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu Province, China
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Hangsheng Zhou
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu Province, China
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Hongyuan Wan
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu Province, China
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Dongjie Yang
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Qing Liu
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
- Huadong Sanatorium, 67 Dajishan, Wuxi 214122, Jiangsu Province, China
| | - Lijie Zhu
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Yuanyuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| |
Collapse
|
5
|
Benz PM, Frömel T, Laban H, Zink J, Ulrich L, Groneberg D, Boon RA, Poley P, Renne T, de Wit C, Fleming I. Cardiovascular Functions of Ena/VASP Proteins: Past, Present and Beyond. Cells 2023; 12:1740. [PMID: 37443774 PMCID: PMC10340426 DOI: 10.3390/cells12131740] [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: 04/26/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Actin binding proteins are of crucial importance for the spatiotemporal regulation of actin cytoskeletal dynamics, thereby mediating a tremendous range of cellular processes. Since their initial discovery more than 30 years ago, the enabled/vasodilator-stimulated phosphoprotein (Ena/VASP) family has evolved as one of the most fascinating and versatile family of actin regulating proteins. The proteins directly enhance actin filament assembly, but they also organize higher order actin networks and link kinase signaling pathways to actin filament assembly. Thereby, Ena/VASP proteins regulate dynamic cellular processes ranging from membrane protrusions and trafficking, and cell-cell and cell-matrix adhesions, to the generation of mechanical tension and contractile force. Important insights have been gained into the physiological functions of Ena/VASP proteins in platelets, leukocytes, endothelial cells, smooth muscle cells and cardiomyocytes. In this review, we summarize the unique and redundant functions of Ena/VASP proteins in cardiovascular cells and discuss the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Peter M. Benz
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
| | - Timo Frömel
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Hebatullah Laban
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Joana Zink
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Lea Ulrich
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Dieter Groneberg
- Institute of Physiology I, University of Würzburg, 97070 Würzburg, Germany
| | - Reinier A. Boon
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
- Cardiopulmonary Institute, 60596 Frankfurt am Main, Germany
- Centre of Molecular Medicine, Institute of Cardiovascular Regeneration, Goethe-University, 60596 Frankfurt am Main, Germany
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Centre, 1081 HZ Amsterdam, The Netherlands
| | - Philip Poley
- Institut für Physiologie, Universität zu Lübeck, 23562 Lübeck, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany
| | - Thomas Renne
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 VN51 Dublin, Ireland
| | - Cor de Wit
- Institut für Physiologie, Universität zu Lübeck, 23562 Lübeck, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
- Cardiopulmonary Institute, 60596 Frankfurt am Main, Germany
| |
Collapse
|
6
|
Li Y, Wang D, Ge H, Güngör C, Gong X, Chen Y. Cytoskeletal and Cytoskeleton-Associated Proteins: Key Regulators of Cancer Stem Cell Properties. Pharmaceuticals (Basel) 2022; 15:1369. [PMID: 36355541 PMCID: PMC9698833 DOI: 10.3390/ph15111369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/02/2022] [Accepted: 11/06/2022] [Indexed: 08/08/2023] Open
Abstract
Cancer stem cells (CSCs) are a subpopulation of cancer cells possessing stemness characteristics that are closely associated with tumor proliferation, recurrence and resistance to therapy. Recent studies have shown that different cytoskeletal components and remodeling processes have a profound impact on the behavior of CSCs. In this review, we outline the different cytoskeletal components regulating the properties of CSCs and discuss current and ongoing therapeutic strategies targeting the cytoskeleton. Given the many challenges currently faced in targeted cancer therapy, a deeper comprehension of the molecular events involved in the interaction of the cytoskeleton and CSCs will help us identify more effective therapeutic strategies to eliminate CSCs and ultimately improve patient survival.
Collapse
Affiliation(s)
- Yuqiang Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Dan Wang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of General Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Heming Ge
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of General Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Cenap Güngör
- Department of General Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Xuejun Gong
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yongheng Chen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
7
|
Zhang Z, Li J, Jiao S, Han G, Zhu J, Liu T. Functional and clinical characteristics of focal adhesion kinases in cancer progression. Front Cell Dev Biol 2022; 10:1040311. [PMID: 36407100 PMCID: PMC9666724 DOI: 10.3389/fcell.2022.1040311] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022] Open
Abstract
Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase and an adaptor protein that primarily regulates adhesion signaling and cell migration. FAK promotes cell survival in response to stress. Increasing evidence has shown that at the pathological level, FAK is highly expressed in multiple tumors in several systems (including lung, liver, gastric, and colorectal cancers) and correlates with tumor aggressiveness and patient prognosis. At the molecular level, FAK promotes tumor progression mainly by altering survival signals, invasive capacity, epithelial-mesenchymal transition, the tumor microenvironment, the Warburg effect, and stemness of tumor cells. Many effective drugs have been developed based on the comprehensive role of FAK in tumor cells. In addition, its potential as a tumor marker cannot be ignored. Here, we discuss the pathological and pre-clinical evidence of the role of FAK in cancer development; we hope that these findings will assist in FAK-based clinical studies.
Collapse
Affiliation(s)
- Zhaoyu Zhang
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jinlong Li
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Simin Jiao
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Guangda Han
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jiaming Zhu
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tianzhou Liu
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
8
|
Zarka M, Haÿ E, Cohen-Solal M. YAP/TAZ in Bone and Cartilage Biology. Front Cell Dev Biol 2022; 9:788773. [PMID: 35059398 PMCID: PMC8764375 DOI: 10.3389/fcell.2021.788773] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/23/2021] [Indexed: 12/25/2022] Open
Abstract
YAP and TAZ were initially described as the main regulators of organ growth during development and more recently implicated in bone biology. YAP and TAZ are regulated by mechanical and cytoskeletal cues that lead to the control of cell fate in response to the cellular microenvironment. The mechanical component represents a major signal for bone tissue adaptation and remodelling, so YAP/TAZ contributes significantly in bone and cartilage homeostasis. Recently, mice and cellular models have been developed to investigate the precise roles of YAP/TAZ in bone and cartilage cells, and which appear to be crucial. This review provides an overview of YAP/TAZ regulation and function, notably providing new insights into the role of YAP/TAZ in bone biology.
Collapse
Affiliation(s)
- Mylène Zarka
- INSERM UMR 1132 BIOSCAR, Hôpital Lariboisière, Paris, France.,Faculté de Santé, Université de Paris, Paris, France
| | - Eric Haÿ
- INSERM UMR 1132 BIOSCAR, Hôpital Lariboisière, Paris, France.,Faculté de Santé, Université de Paris, Paris, France
| | - Martine Cohen-Solal
- INSERM UMR 1132 BIOSCAR, Hôpital Lariboisière, Paris, France.,Faculté de Santé, Université de Paris, Paris, France
| |
Collapse
|
9
|
Lim YX, Lin H, Seah SH, Lim YP. Reciprocal Regulation of Hippo and WBP2 Signalling-Implications in Cancer Therapy. Cells 2021; 10:cells10113130. [PMID: 34831354 PMCID: PMC8625973 DOI: 10.3390/cells10113130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/08/2021] [Accepted: 11/08/2021] [Indexed: 11/23/2022] Open
Abstract
Cancer is a global health problem. The delineation of molecular mechanisms pertinent to cancer initiation and development has spurred cancer therapy in the form of precision medicine. The Hippo signalling pathway is a tumour suppressor pathway implicated in a multitude of cancers. Elucidation of the Hippo pathway has revealed an increasing number of regulators that are implicated, some being potential therapeutic targets for cancer interventions. WW domain-binding protein 2 (WBP2) is an oncogenic transcriptional co-factor that interacts, amongst others, with two other transcriptional co-activators, YAP and TAZ, in the Hippo pathway. WBP2 was recently discovered to modulate the upstream Hippo signalling components by associating with LATS2 and WWC3. Exacerbating the complexity of the WBP2/Hippo network, WBP2 itself is reciprocally regulated by Hippo-mediated microRNA biogenesis, contributing to a positive feedback loop that further drives carcinogenesis. Here, we summarise the biological mechanisms of WBP2/Hippo reciprocal regulation and propose therapeutic strategies to overcome Hippo defects in cancers through targeting WBP2.
Collapse
Affiliation(s)
- Yvonne Xinyi Lim
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore 119077, Singapore; (Y.X.L.); (H.L.); (S.H.S.)
- Department of Biochemistry, National University of Singapore, Singapore 117596, Singapore
| | - Hexian Lin
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore 119077, Singapore; (Y.X.L.); (H.L.); (S.H.S.)
- Department of Biochemistry, National University of Singapore, Singapore 117596, Singapore
| | - Sock Hong Seah
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore 119077, Singapore; (Y.X.L.); (H.L.); (S.H.S.)
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Yoon Pin Lim
- Department of Biochemistry, National University of Singapore, Singapore 117596, Singapore
- Correspondence:
| |
Collapse
|
10
|
Cinar B, Alp E, Al-Mathkour M, Boston A, Dwead A, Khazaw K, Gregory A. The Hippo pathway: an emerging role in urologic cancers. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2021; 9:301-317. [PMID: 34541029 PMCID: PMC8446764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 08/25/2021] [Indexed: 06/13/2023]
Abstract
The Hippo pathway controls several biological processes, including cell growth, differentiation, motility, stemness, cell contact, immune cell maturation, organ size, and tumorigenesis. The Hippo pathway core kinases MST1/2 and LATS1/2 in mammals phosphorylate and inactivate YAP1 signaling. Increasing evidence indicates that loss of MST1/2 and LATS1/2 function is linked to the biology of many cancer types with poorer outcomes, likely due to the activation of oncogenic YAP1/TEAD signaling. Therefore, there is a renewed interest in blocking the YAP1/TEAD functions to prevent cancer growth. This review introduces the Hippo pathway components and examines their role and therapeutic potentials in prostate, kidney, and bladder cancer.
Collapse
Affiliation(s)
- Bekir Cinar
- The Center for Cancer Research and Therapeutic Development, Department of Biological Sciences, Clark Atlanta University Atlanta, Georgia, USA
| | - Esma Alp
- The Center for Cancer Research and Therapeutic Development, Department of Biological Sciences, Clark Atlanta University Atlanta, Georgia, USA
| | - Marwah Al-Mathkour
- The Center for Cancer Research and Therapeutic Development, Department of Biological Sciences, Clark Atlanta University Atlanta, Georgia, USA
| | - Ava Boston
- The Center for Cancer Research and Therapeutic Development, Department of Biological Sciences, Clark Atlanta University Atlanta, Georgia, USA
| | - Abdulrahman Dwead
- The Center for Cancer Research and Therapeutic Development, Department of Biological Sciences, Clark Atlanta University Atlanta, Georgia, USA
| | - Kezhan Khazaw
- The Center for Cancer Research and Therapeutic Development, Department of Biological Sciences, Clark Atlanta University Atlanta, Georgia, USA
| | - Alexis Gregory
- The Center for Cancer Research and Therapeutic Development, Department of Biological Sciences, Clark Atlanta University Atlanta, Georgia, USA
| |
Collapse
|
11
|
Chuo D, Lin D, Yin M, Chen Y. Genetic Variants of the MIF Gene and Susceptibility of Rectal Cancer. Pharmgenomics Pers Med 2021; 14:55-60. [PMID: 33469341 PMCID: PMC7812028 DOI: 10.2147/pgpm.s282653] [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: 09/18/2020] [Accepted: 12/05/2020] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Rectal cancer (RC) has been documented to be a highly invasive malignant neoplasm worldwide. Macrophage migration inhibitory factor (MIF) is a multifunctional cytokine involved in cell-mediated immunity, immunoregulation, inflammation. In vitro and in vivo studies have identified that MIF was involved in the carcinogenesis and progression of RC. PATIENTS AND METHODS This case-control study evaluated associations of genetic variants of the MIF gene and serum level of MIF with susceptibility of RC. RESULTS We found MIF level was associated with an increased risk of RC (OR for per unit: 1.38, 95% CI:1.32-1.44; P < 0.001). Both MIF rs2012133 (OR = 1.30; 95% CIs = 1.08-1.58; P = 0.007) and rs755622 (OR = 1.45; 95% CIs = 1.15-1.82; P = 0.002) were significantly associated with increased risk of RC. Besides, we also found MIF rs5844572 was significantly associated with increased susceptibility of RC, with OR for per CATT repeat of 1.28 (95% CIs: 1.16-1.41; P < 0.001). Further, we found all three variants of the MIF gene, rs5844572, rs2012133 and rs755622, could increase serum level of MIF. CONCLUSION This study suggests that MIF plays an important role in the carcinogenesis of RC and could be used as a biomarker for early detection and prediction of RC.
Collapse
Affiliation(s)
- Dongyu Chuo
- Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province110042, People’s Republic of China
| | - Dapeng Lin
- Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province110042, People’s Republic of China
| | - Mingdi Yin
- Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province110042, People’s Republic of China
| | - Yuze Chen
- Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province110042, People’s Republic of China
| |
Collapse
|
12
|
Association between Inflammation and Function of Cell Adhesion Molecules Influence on Gastrointestinal Cancer Development. Cells 2021; 10:cells10010067. [PMID: 33406733 PMCID: PMC7824562 DOI: 10.3390/cells10010067] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/09/2020] [Accepted: 12/29/2020] [Indexed: 12/16/2022] Open
Abstract
Gastrointestinal cancer is highly associated with inflammatory processes inducing the release of cytokines from cancer or immune cells, including interferons, interleukins, chemokines, colony-stimulating factors, and growth factors, which promote or suppress tumor progression. Inflammatory cytokines within the tumor microenvironment promote immune cell infiltration. Infiltrating immune, and tumor-surrounding stromal cells support tumor growth, angiogenesis, metastasis, and immunosuppression through communication with inflammatory cytokines and cell adhesion molecules. Notably, infiltrating immune and tumor cells present immunosuppressive molecules, such as programmed death-ligand 1 (PD-L1) and CD80/CD86. Suppression of cytotoxic T cells promotes tumor avoidance of immune surveillance and greater malignancy. Moreover, glycosylation and sialylation of proteins hyperexpressed on the cancer cell surface have been shown to enhance immune escape and metastasis. Cytokine treatments and immune checkpoint inhibitors are widely used in clinical practice. However, the tumor microenvironment is a rapidly changing milieu involving several factors. In this review, we have provided a summary of the interactions of inflammation and cell adhesion molecules between cancer and other cell types, to improve understanding of the tumor microenvironment.
Collapse
|
13
|
Moss AC, Herr AE. In-gel fluorescence detection by DNA polymerase elongation. APL Bioeng 2020; 4:046104. [PMID: 33263097 PMCID: PMC7680656 DOI: 10.1063/5.0021149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/07/2020] [Indexed: 11/14/2022] Open
Abstract
Fluorescence-based DNA readouts are increasingly important in biological research, owing to enhanced analytical sensitivity and multiplexing capability. In this study, we characterize an in-gel polymerase elongation process to understand the reaction kinetics and transport limitations, and we evaluate DNA sequence design to develop signal amplification strategies. Using fluorescently labeled nucleotides, we scrutinize polymerase elongation on single-stranded overhangs of DNA immobilized in polyacrylamide hydrogels. When polymerase elongation reactions were carried out with reactants diffused into the gels, we observed reaction completion after 2 h, indicating that the process was efficient but much slower than that predicted by models. Confocal microscopy revealed a nonuniform post-reaction fluorescence profile of the elongated DNA throughout the depth of the gel and that the time for complete fluorescence penetration was proportional to the immobilized DNA concentration. These observations suggest retarded diffusion of the polymerase, attributable to interactions between diffusing polymerase and immobilized DNA. This study will ultimately inform assay design by providing insight into the reaction completion time to ensure spatial uniformity of the fluorescence signal. In agreement with our hypothesis that incorporation of multiple labeled nucleotides per DNA strand results in an increased signal, incorporation of four labeled nucleotides resulted in a 2.3-fold increase in fluorescence intensity over one labeled nucleotide. Our results further suggest that the fluorescence signal increases with spacing between labeled nucleotides, validating the number of and spacing between labeled nucleotides as tunable parameters for signal amplification. In-gel polymerase-based fluorescence readout is promising for signal amplification when considering both transport limitations and DNA sequence design.
Collapse
Affiliation(s)
| | - Amy E. Herr
- Author to whom correspondence should be addressed:
| |
Collapse
|
14
|
Abstract
Simple Summary Cell migration is an essential process from embryogenesis to cell death. This is tightly regulated by numerous proteins that help in proper functioning of the cell. In diseases like cancer, this process is deregulated and helps in the dissemination of tumor cells from the primary site to secondary sites initiating the process of metastasis. For metastasis to be efficient, cytoskeletal components like actin, myosin, and intermediate filaments and their associated proteins should co-ordinate in an orderly fashion leading to the formation of many cellular protrusions-like lamellipodia and filopodia and invadopodia. Knowledge of this process is the key to control metastasis of cancer cells that leads to death in 90% of the patients. The focus of this review is giving an overall understanding of these process, concentrating on the changes in protein association and regulation and how the tumor cells use it to their advantage. Since the expression of cytoskeletal proteins can be directly related to the degree of malignancy, knowledge about these proteins will provide powerful tools to improve both cancer prognosis and treatment. Abstract Successful metastasis depends on cell invasion, migration, host immune escape, extravasation, and angiogenesis. The process of cell invasion and migration relies on the dynamic changes taking place in the cytoskeletal components; actin, tubulin and intermediate filaments. This is possible due to the plasticity of the cytoskeleton and coordinated action of all the three, is crucial for the process of metastasis from the primary site. Changes in cellular architecture by internal clues will affect the cell functions leading to the formation of different protrusions like lamellipodia, filopodia, and invadopodia that help in cell migration eventually leading to metastasis, which is life threatening than the formation of neoplasms. Understanding the signaling mechanisms involved, will give a better insight of the changes during metastasis, which will eventually help targeting proteins for treatment resulting in reduced mortality and longer survival.
Collapse
|
15
|
Chen C, Wang Y, Chen S, Ruan X, Liao H, Zhang Y, Sun J, Gao J, Deng G. Genistein inhibits migration and invasion of cervical cancer HeLa cells by regulating FAK-paxillin and MAPK signaling pathways. Taiwan J Obstet Gynecol 2020; 59:403-408. [DOI: 10.1016/j.tjog.2020.03.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2020] [Indexed: 01/14/2023] Open
|
16
|
TAZ target gene ITGAV regulates invasion and feeds back positively on YAP and TAZ in liver cancer cells. Cancer Lett 2020; 473:164-175. [PMID: 31904487 DOI: 10.1016/j.canlet.2019.12.044] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/13/2019] [Accepted: 12/25/2019] [Indexed: 12/13/2022]
Abstract
The Hippo pathway effectors yes-associated protein (YAP) and WW domain containing transcription regulator 1 (TAZ/WWTR1) support tumor initiation and progression in various cancer entities including hepatocellular carcinoma (HCC). However, to which extent YAP and TAZ contribute to liver tumorigenesis via common and exclusive molecular mechanisms is poorly understood. RNAinterference (RNAi) experiments illustrate that YAP and TAZ individually support HCC cell viability and migration, while for invasion additive effects were observed. Comprehensive expression profiling revealed partly overlapping YAP/TAZ target genes as well as exclusively regulated genes. Integrin-αV (ITGAV) is a novel TAZ-specific target gene, whose overexpression in human HCC patients correlates with poor clinical outcome, TAZ expression in HCCs, and the abundance of YAP/TAZ target genes. Functionally, ITGAV contributes to actin stress fiber assembly, tumor cell migration and invasion. Perturbation of ITGAV diminishes actin fiber formation and nuclear YAP/TAZ protein levels. We describe a novel Hippo downstream mechanism in HCC cells, which is regulated by TAZ and ITGAV and that feedbacks on YAP/TAZ activity. This mechanism may represent a therapeutic target structure since it contributes to signal amplification of oncogenic YAP/TAZ in hepatocarcinogenesis.
Collapse
|
17
|
Zhou B, Zong S, Zhong W, Tian Y, Wang L, Zhang Q, Zhang R, Li L, Wang W, Zhao J, Chen X, Feng Y, Zhai B, Sun T, Liu Y. Interaction between laminin-5γ2 and integrin β1 promotes the tumor budding of colorectal cancer via the activation of Yes-associated proteins. Oncogene 2019; 39:1527-1542. [PMID: 31676872 DOI: 10.1038/s41388-019-1082-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is a common cancer type and a threat to human health. Tumor budding (TB) is the presence of a single cancer cell or clusters of up to five cancer cells prior to the invasive front of an aggressive carcinoma and is an independent prognosis factor for CRC. The molecular mechanism of TB is still unclear, and drugs that inhibit this process are still in the blank stage. This study found that TBs exhibit characteristics of partial EMT with a decreased expression of E-cadherin and no substantial differences in the expression of N-cadherin and vimentin. We also observed the interaction of integrin with extracellular matrix components, laminin-5γ2 (LN-5γ2), play essential roles in the TB of CRC. We then verified that the interaction between LN-5γ2 and integrin β1 promotes the TB of CRC via the activation of FAK and Yes-associated proteins (YAP). A natural drug monomer, cucurbitacin B, was screened using virtual screening methods for the interaction interface of proteins. We found that this monomer could block the interaction interface between LN-5γ2 and integrin β1 and substantially inhibit the TB of CRC cells via inactivation of YAP. This study provides new insights into the mechanism of TB mechanism and the development of drugs targeting the TB of CRC.
Collapse
Affiliation(s)
- Bijiao Zhou
- Molecular Pathology Institute of Gastrointestinal Tumors, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China.,State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China.,Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China
| | - Shumin Zong
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China.,Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Yixuan Tian
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China.,Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China
| | - Lumeng Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China.,Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China
| | - Qian Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China.,Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China
| | - Renya Zhang
- Molecular Pathology Institute of Gastrointestinal Tumors, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China
| | - Lei Li
- Molecular Pathology Institute of Gastrointestinal Tumors, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China
| | - Wei Wang
- Molecular Pathology Institute of Gastrointestinal Tumors, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China
| | - Jianmin Zhao
- Department of Pathology, Hospital of Shun Yi District, Beijing, China
| | - Xin Chen
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China.,Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China
| | - Yaju Feng
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China.,Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China
| | - Binghui Zhai
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China.,Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China
| | - Tao Sun
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China. .,Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China.
| | - Yanrong Liu
- Molecular Pathology Institute of Gastrointestinal Tumors, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China. .,State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China. .,Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China.
| |
Collapse
|
18
|
Wang Y, Tu K, Liu D, Guo L, Chen Y, Li Q, Maiers JL, Liu Z, Shah VH, Dou C, Tschumperlin D, Voneschen L, Yang R, Kang N. p300 Acetyltransferase Is a Cytoplasm-to-Nucleus Shuttle for SMAD2/3 and TAZ Nuclear Transport in Transforming Growth Factor β-Stimulated Hepatic Stellate Cells. Hepatology 2019; 70:1409-1423. [PMID: 31004519 PMCID: PMC6783326 DOI: 10.1002/hep.30668] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 04/11/2019] [Indexed: 12/12/2022]
Abstract
Nuclear translocation of mothers against decapentaplegic homolog 2/3 (SMAD2/3), core transcription factors of transforming growth factor β (TGF-β) signaling, is critical for hepatic stellate cell (HSC) differentiation into metastasis-promoting myofibroblasts. SMAD2/3 have multiple coactivators, including WW domain-containing transcription regulator protein 1 (WWTR1 or TAZ) and p300 acetyltransferase. In the nucleus, TAZ binds to SMAD2/3 to prevent SMAD2/3 nuclear export. However, how TAZ and SMAD2/3 enter the nucleus remains poorly understood because neither contains a nuclear localization signal (NLS), an amino acid sequence tagging proteins for nuclear transport. p300 is an NLS-containing large scaffold protein, so we hypothesized that SMAD2/3 and TAZ may undergo nuclear import through complexing with p300. Coimmunoprecipitation, immunofluorescence, and nuclear fractionation assays revealed that TGF-β1 promoted binding of SMAD2/3 and TAZ to p300 and that p300 inactivation disrupted TGF-β1-mediated SMAD2/3 and TAZ nuclear accumulation. Deleting the p300 NLS blocked TGF-β1-induced SMAD2/3 and TAZ nuclear transport. Consistently, p300 inactivation suppressed TGF-β1-mediated HSC activation and transcription of genes encoding tumor-promoting factors, such as connective tissue growth factor, Tenascin C, Periostin, platelet-derived growth factor C, and fibroblast growth factor 2, as revealed by microarray analysis. Chromatin immunoprecipitation-real-time quantitative PCR showed that canonical p300-mediated acetylation of histones also facilitated transcription in response to TGF-β1 stimulation. Interestingly, although both TGF-β1-mediated and stiffness-mediated HSC activation require p300, comparison of gene expression data sets revealed that transcriptional targets of TGF-β1 were distinct from those of stiffness-p300 mechanosignaling. Lastly, in tumor/HSC coinjection and intrasplenic tumor injection models, targeting p300 of activated-HSC/myofibroblasts by C646, short hairpin RNA, or cre-mediated gene disruption reduced tumor and liver metastatic growth in mice. Conclusion: p300 facilitates TGF-β1-stimulated HSC activation by both noncanonical (cytoplasm-to-nucleus shuttle for SMAD2/3 and TAZ) and canonical (histone acetylation) mechanisms. p300 is an attractive target for inhibiting HSC activation and the prometastatic liver microenvironment.
Collapse
Affiliation(s)
- Yuanguo Wang
- Tumor Microenvironment and MetastasisHormel Institute, University of MinnesotaAustinMN
| | - Kangsheng Tu
- Tumor Microenvironment and MetastasisHormel Institute, University of MinnesotaAustinMN
- 1st Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShanxiP. R. China
| | - Donglian Liu
- Tumor Microenvironment and MetastasisHormel Institute, University of MinnesotaAustinMN
- 6th Affiliated Hospital of Guangzhou Medical UniversityQingyuanGuangdongP. R. China
| | - Luyang Guo
- Tumor Microenvironment and MetastasisHormel Institute, University of MinnesotaAustinMN
| | - Yunru Chen
- Tumor Microenvironment and MetastasisHormel Institute, University of MinnesotaAustinMN
- 1st Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShanxiP. R. China
| | - Qing Li
- Tumor Microenvironment and MetastasisHormel Institute, University of MinnesotaAustinMN
- 1st Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShanxiP. R. China
| | - Jessica L. Maiers
- GI Research Unit and Cancer Cell Biology ProgramMayo ClinicRochesterMN
| | - Zhikui Liu
- 1st Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShanxiP. R. China
- GI Research Unit and Cancer Cell Biology ProgramMayo ClinicRochesterMN
| | - Vijay H. Shah
- GI Research Unit and Cancer Cell Biology ProgramMayo ClinicRochesterMN
| | - Changwei Dou
- GI Research Unit and Cancer Cell Biology ProgramMayo ClinicRochesterMN
- Zhejiang Provincial People’s HospitalHangzhouZhejiangP. R. China
| | | | - Luke Voneschen
- Computational Cancer GenomicsHormel Institute, University of MinnesotaAustinMN
| | - Rendong Yang
- Computational Cancer GenomicsHormel Institute, University of MinnesotaAustinMN
| | - Ningling Kang
- Tumor Microenvironment and MetastasisHormel Institute, University of MinnesotaAustinMN
| |
Collapse
|
19
|
Xu S, Zhang H, Chong Y, Guan B, Guo P. YAP Promotes VEGFA Expression and Tumor Angiogenesis Though Gli2 in Human Renal Cell Carcinoma. Arch Med Res 2019; 50:225-233. [PMID: 31518897 DOI: 10.1016/j.arcmed.2019.08.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/30/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND High vascularization is a major characteristic of renal cell carcinoma (RCC). Thus, exploration of molecules promoting the tumor vascularization in RCC is urgent. Yes-associated Protein (YAP) is an oncogene in many cancer types, and high YAP expression was correlated with worse overall survival of RCC patients according to The Cancer Genome Atlas (TCGA) database. However, whether YAP promotes tumor angiogenesis of RCC is still unknown. METHODS Western blotting assay, real-time Quantitive PCR analysis, and ELISA assay were used to detect the related gene expression. The function of YAP on tumor angiogenesis was investigated by HUVEC recruitment, tube formation, and rabbit cornea assay. The clinical relevance of several genes was analyzed in a public database. RESULTS knockdown of YAP decreased RCC cell-inducing HUVEC recruitment and tube formation. Moreover, tumor angiogenesis ability of 786-O cells was crippled by YAP knockdown in vivo. In addition, the expression of Vascular endothelial growth factors A (VEGFA) was positively correlated with YAP expression in RCC tumor tissues, and YAP promoted expression and secretion of VEGFA in RCC cells. Mechanistically, GLI family zinc finger 2 (Gli2) knockdown in RCC cells reduced both basic and YAP-induced VEGFA expression, HUVECs recruitment, and tube formation, indicating that Gli2 is necessary for YAP to promote expression of VEGFA. CONCLUSION Taken together, our results demonstrate that YAP/Gli2 promotes VEGFA expression and tumor angiogenesis in RCC cells, which could provide novel therapeutic targets in RCC treatment.
Collapse
Affiliation(s)
- Shan Xu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Haibao Zhang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Yue Chong
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Bing Guan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Peng Guo
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China.
| |
Collapse
|
20
|
Lai YC, Huang KH, Chen MH, Chao Y, Lo SS, Li AFY, Wu CW, Shyr YM, Fang WL. The Clinical Implication of PTEN and FAK Expression in Gastric Cancer Patients. Int Surg 2019; 104:48-57. [DOI: 10.9738/intsurg-d-19-00021.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
Objective:
The tumor suppressor gene phosphatase and tensin homolog (PTEN) was reported to inhibit the growth and invasion of gastric cancer (GC) via the downregulation of focal adhesion kinase (FAK). To date, the clinical implication of PTEN and FAK expression in GC has not been well addressed.
Methods:
A total of 200 GC patients receiving curative surgery were enrolled. The clinicopathologic features according to the expression of PTEN and FAK protein using immunohistochemical staining were compared among patients.
Results:
Patients with high PTEN expression were more likely to have smaller tumor size, more well- and moderately differentiated tumors, a more superficial gross appearance, less scirrhous stromal reactions, more likely to have high FAK expression, and have less advanced pathologic tumor (T) category, node (N) category, and tumor, node, metastasis (TNM) stage and more distant metastases than patients with low PTEN expression. Multivariate analysis showed that PTEN/FAK expression status is an independent prognostic factor affecting overall survival (OS) and disease-free survival (DFS). Patients with PTEN(high)/FAK(low) had better OS and DFS, followed by those with PTEN(high)/FAK(high), those with PTEN(low)/FAK(low), and those with PTEN(low)/FAK(high) (OS: 83.3% versus 58.0% versus 46.2% versus 26.5%, respectively, P < 0.001; DFS: 83.3% versus 55.8% versus 30.8% versus 24.4%, respectively, P < 0.001).
Conclusions:
GC patients with high PTEN expression were more likely to have fewer tumor recurrences and a better prognosis than those with low PTEN expression. PTEN and FAK may have opposing effects on GC patient survival. Our results may have clinical impact on treatment of GC patients.
Collapse
Affiliation(s)
- Yi-Chen Lai
- Department of Radiology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Kuo-Hung Huang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Huang Chen
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Center of Immuno-Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yee Chao
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Center of Immuno-Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Su-Shun Lo
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- National Yang-Ming University Hospital, Yilan, Taiwan
| | - Anna Fen-Yau Li
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Pathology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chew-Wun Wu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Ming Shyr
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wen-Liang Fang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|