1
|
Wang W, Zhu L, Zhou J, Liu X, Xiao M, Chen N, Huang X, Chen H, Pei X, Zhang H. Targeting the KRT16-vimentin axis for metastasis in lung cancer. Pharmacol Res 2023:106818. [PMID: 37315823 DOI: 10.1016/j.phrs.2023.106818] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 05/23/2023] [Accepted: 06/08/2023] [Indexed: 06/16/2023]
Abstract
Lung cancer is the most diagnosed malignant cancer and the leading cause of cancer-related deaths worldwide, with advanced stage and metastasis being a major issue. The mechanism leading to metastasis is not yet understood. Here, we found that KRT16 is upregulated in metastatic lung cancer tissues and correlated with poor overall survival. Knockdown of KRT16 inhibits metastasis of lung cancer both in vitro and in vivo. Mechanistically, KRT16 interacts with vimentin, and depletion of KRT16 leads to downregulation of vimentin. KRT16 acquired its oncogenic ability by stabilizing vimentin, and vimentin is required for KRT16-driven metastasis. FBXO21 mediates the polyubiquitination and degradation of KRT16, and vimentin inhibits KRT16 ubiquitination and degradation by impairing its interaction with FBXO21. Significantly, IL-15 inhibits metastasis of lung cancer in a mouse model through upregulation of FBXO21, and the level of IL-15 in circulating serum was significantly higher in nonmetastatic lung cancer patients than in metastatic patients. Our findings indicate that targeting the FBXO21/KRT16/vimentin axis may benefit lung cancer patients with metastasis.
Collapse
Affiliation(s)
- Wen Wang
- Department of Oncology, The fifth Affiliated Hospital of Sun Yat-sen University, 519000(,) Zhuhai(,) China.
| | - Lifei Zhu
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, 510515(,) Guangzhou(,) China.
| | - Jiao Zhou
- Department of Oncology, The fifth Affiliated Hospital of Sun Yat-sen University, 519000(,) Zhuhai(,) China.
| | - Xiaoli Liu
- Department of Oncology, The fifth Affiliated Hospital of Sun Yat-sen University, 519000(,) Zhuhai(,) China.
| | - Mei Xiao
- Department of Oncology, The fifth Affiliated Hospital of Sun Yat-sen University, 519000(,) Zhuhai(,) China.
| | - Nan Chen
- Department of Oncology, The fifth Affiliated Hospital of Sun Yat-sen University, 519000(,) Zhuhai(,) China.
| | - Xiaodan Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.
| | - Hongtao Chen
- Department of Laboratory, The fifth Affiliated Hospital of Sun Yat-sen University, 519000(,) Zhuhai(,) China.
| | - Xiaofeng Pei
- Department of Oncology, The fifth Affiliated Hospital of Sun Yat-sen University, 519000(,) Zhuhai(,) China.
| | - Hongyu Zhang
- Department of Oncology, The fifth Affiliated Hospital of Sun Yat-sen University, 519000(,) Zhuhai(,) China; Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Zhuhai, China.
| |
Collapse
|
2
|
Rademaker E, Bastiaannet E, Oosting J, Dekker-Ensink NG, Kuppen PJK, de Miranda NFCC, Liefers GJ. Revising the Role of Integrin Subunit β4 Expression in Colon Cancer Progression and Survival. J Gastrointest Cancer 2023; 54:147-154. [PMID: 35112314 PMCID: PMC10182939 DOI: 10.1007/s12029-021-00787-8] [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] [Accepted: 12/06/2021] [Indexed: 11/29/2022]
Abstract
PURPOSE Integrin subunit β4 (β4) has been proposed to play an important role in colon cancer progression through its involvement in hemidesmosome disassembly processes and tumor cell migration. However, the association between β4 expression and clinicopathological outcomes in colon cancer remains unclear. METHODS Expression of β4 was assessed by immunohistochemistry in a large cohort of 651 colon cancer patients, the largest colon cancer cohort so far. Chi-squared tests were used to study the association between β4 expression and clinicopathological features. Overall and disease-free survival were assessed by Cox proportional hazard models. RESULTS Loss of β4 expression was associated with local tumor invasion. Only 17.9% of the pT1 tumors displayed weak β4 expression level versus 28.1% of pT4 tumors, and 25.0% of the pT1 tumors had a high expression level versus 8.6% of the pT4 tumors (p = 0.012). No association between β4 expression and overall (p = 0.845) or disease-free survival (p = 0.767) was encountered, which disputes the role of β4 as a biomarker of malignant behavior in colon cancer. CONCLUSION Contradictory reports have suggested opposite roles for β4 expression in (colon) cancer progression. In the present large cohort of colon cancer patients, we found that β4 expression was not associated with worse clinical prognosis, but decreased with advanced pathological tumor stage. Future studies should establish whether loss of β4 expression promotes invasive characteristics of colon cancer cells.
Collapse
Affiliation(s)
- Eva Rademaker
- Department of Surgical Oncology, Leiden University Medical Center, P.O. Box 9600, Leiden, 2300 RC, The Netherlands.
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Esther Bastiaannet
- Department of Surgical Oncology, Leiden University Medical Center, P.O. Box 9600, Leiden, 2300 RC, The Netherlands
| | - Jan Oosting
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Neeltje G Dekker-Ensink
- Department of Surgical Oncology, Leiden University Medical Center, P.O. Box 9600, Leiden, 2300 RC, The Netherlands
| | - Peter J K Kuppen
- Department of Surgical Oncology, Leiden University Medical Center, P.O. Box 9600, Leiden, 2300 RC, The Netherlands
| | | | - Gerrit J Liefers
- Department of Surgical Oncology, Leiden University Medical Center, P.O. Box 9600, Leiden, 2300 RC, The Netherlands
| |
Collapse
|
3
|
Wang EJY, Chen IH, Kuo BYT, Yu CC, Lai MT, Lin JT, Lin LYT, Chen CM, Hwang T, Sheu JJC. Alterations of Cytoskeleton Networks in Cell Fate Determination and Cancer Development. Biomolecules 2022; 12:biom12121862. [PMID: 36551290 PMCID: PMC9775460 DOI: 10.3390/biom12121862] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/03/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022] Open
Abstract
Cytoskeleton proteins have been long recognized as structural proteins that provide the necessary mechanical architecture for cell development and tissue homeostasis. With the completion of the cancer genome project, scientists were surprised to learn that huge numbers of mutated genes are annotated as cytoskeletal or associated proteins. Although most of these mutations are considered as passenger mutations during cancer development and evolution, some genes show high mutation rates that can even determine clinical outcomes. In addition, (phospho)proteomics study confirms that many cytoskeleton-associated proteins, e.g., β-catenin, PIK3CA, and MB21D2, are important signaling mediators, further suggesting their biofunctional roles in cancer development. With emerging evidence to indicate the involvement of mechanotransduction in stemness formation and cell differentiation, mutations in these key cytoskeleton components may change the physical/mechanical properties of the cells and determine the cell fate during cancer development. In particular, tumor microenvironment remodeling triggered by such alterations has been known to play important roles in autophagy, metabolism, cancer dormancy, and immune evasion. In this review paper, we will highlight the current understanding of how aberrant cytoskeleton networks affect cancer behaviors and cellular functions through mechanotransduction.
Collapse
Affiliation(s)
- Evan Ja-Yang Wang
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - I-Hsuan Chen
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813405, Taiwan
- Department of Pharmacy, College of Pharmacy and Health Care, Tajen University, Pingtung County 907391, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Brian Yu-Ting Kuo
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - Chia-Cheng Yu
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813405, Taiwan
- Department of Pharmacy, College of Pharmacy and Health Care, Tajen University, Pingtung County 907391, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| | - Ming-Tsung Lai
- Department of Pathology, Taichung Hospital, Ministry of Health and Welfare, Taichung 403301, Taiwan
| | - Jen-Tai Lin
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813405, Taiwan
| | - Leo Yen-Ting Lin
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - Chih-Mei Chen
- Human Genetic Center, China Medical University Hospital, Taichung 404327, Taiwan
| | - Tritium Hwang
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - Jim Jinn-Chyuan Sheu
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
- Institute of Precision Medicine, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
- Correspondence: ; Tel.: +886-7-5252000 (ext. 7102)
| |
Collapse
|
4
|
Fischer NG, Aparicio C. Junctional epithelium and hemidesmosomes: Tape and rivets for solving the "percutaneous device dilemma" in dental and other permanent implants. Bioact Mater 2022; 18:178-198. [PMID: 35387164 PMCID: PMC8961425 DOI: 10.1016/j.bioactmat.2022.03.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/14/2022] [Accepted: 03/12/2022] [Indexed: 02/06/2023] Open
Abstract
The percutaneous device dilemma describes etiological factors, centered around the disrupted epithelial tissue surrounding non-remodelable devices, that contribute to rampant percutaneous device infection. Natural percutaneous organs, in particular their extracellular matrix mediating the "device"/epithelium interface, serve as exquisite examples to inspire longer lasting long-term percutaneous device design. For example, the tooth's imperviousness to infection is mediated by the epithelium directly surrounding it, the junctional epithelium (JE). The hallmark feature of JE is formation of hemidesmosomes, cell/matrix adhesive structures that attach surrounding oral gingiva to the tooth's enamel through a basement membrane. Here, the authors survey the multifaceted functions of the JE, emphasizing the role of the matrix, with a particular focus on hemidesmosomes and their five main components. The authors highlight the known (and unknown) effects dental implant - as a model percutaneous device - placement has on JE regeneration and synthesize this information for application to other percutaneous devices. The authors conclude with a summary of bioengineering strategies aimed at solving the percutaneous device dilemma and invigorating greater collaboration between clinicians, bioengineers, and matrix biologists.
Collapse
Affiliation(s)
- Nicholas G. Fischer
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, MN, 55455, USA
| | - Conrado Aparicio
- MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-212 Moos Tower, 515 Delaware St. SE, Minneapolis, MN, 55455, USA
- Division of Basic Research, Faculty of Odontology, UIC Barcelona – Universitat Internacional de Catalunya, C/. Josep Trueta s/n, 08195, Sant Cugat del Valles, Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), C/. Baldiri Reixac 10-12, 08028, Barcelona, Spain
| |
Collapse
|
5
|
Signaling Pathways Associated with Chronic Wound Progression: A Systems Biology Approach. Antioxidants (Basel) 2022; 11:antiox11081506. [PMID: 36009225 PMCID: PMC9404828 DOI: 10.3390/antiox11081506] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 11/23/2022] Open
Abstract
Previously we have shown that several oxidative stress-driven pathways in cutaneous chronic wounds are dysregulated in the first 48 h post-wounding. Here, we performed an RNASeq analysis of tissues collected up to day 20 after wounding, when we have determined full chronicity is established. Weighted Gene Correlation Network Analysis was performed in R segregating the genes into 14 modules. Genes in the modules significantly correlated (p < 0.05) to early and full chronicity were used for pathway analysis using pathfindR. In early chronicity, we observed enrichment of several pathways. Dysregulation of Ephrin/Eph signaling leads to growth cone collapse and impairs neuronal regeneration. Adra2b and Adra2a overexpression in early and full chronicity, respectively, decreased cAMP production and impaired re-epithelialization and granulation tissue formation. Several pathways involving a Smooth-muscle-actin (Acta1) were also enriched with Acta1 overexpression contributing to impaired angiogenesis. During full chronicity, the ‘JAK-STAT’ pathway was suppressed undermining host defenses against infection. Wnt signaling was also suppressed, impairing re-epithelialization and granulation tissue formation. Biomarkers of cancer such as overexpression of SDC1 and constitutive activation of ErbB2/HER2 were also identified. In conclusion, we show that during progression to full chronicity, numerous signaling pathways are dysregulated, including some related to carcinogenesis, suggesting that chronic wounds behave much like cancer. Experimental verification in vivo could identify candidates for treatment of chronic wounds.
Collapse
|
6
|
Hou S, Hao X, Li J, Weng S, Wang J, Zhao T, Li W, Hu X, Deng B, Gu J, Hang Q. TM4SF1 promotes esophageal squamous cell carcinoma metastasis by interacting with integrin α6. Cell Death Dis 2022; 13:609. [PMID: 35835740 PMCID: PMC9283456 DOI: 10.1038/s41419-022-05067-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 07/03/2022] [Accepted: 07/04/2022] [Indexed: 01/21/2023]
Abstract
Transmembrane-4 L-six family member-1 (TM4SF1) is a member of the L6 family and functions as a signal transducer to regulate tumor cell behaviors. However, the function and mechanism of TM4SF1 in esophageal squamous cell carcinoma (ESCC) metastasis remains unclear. Here, we find that TM4SF1 expression is increased and positively correlated with clinical TNM stage, N classification, differentiation, tumor size, and poor prognosis in ESCC patients. Interestingly, we demonstrate that TM4SF1 promotes ESCC cell adhesion, spreading, migration, and invasion, but not cell proliferation, in a laminin-dependent manner by interacting with integrin α6. Mechanistically, the TM4SF1/integrin α6/FAK axis signal pathway mediates cell migration under laminin-coating condition. Inhibiting FAK or knocking down TM4SF1 can attenuate TM4SF1-mediated cell migration and lung metastasis. Clinically, the TM4SF1/integrin α6/FAK axis positively correlates with ESCC. Altogether, these findings reveal a new mechanism of TM4SF1 in promoting ESCC metastasis via binding to integrin α6 and suggest that the cross-talk between TM4SF1 and integrin α6 may serve as a therapeutic target for ESCC.
Collapse
Affiliation(s)
- Sicong Hou
- grid.452743.30000 0004 1788 4869Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, 225009 Yangzhou, Jiangsu China
| | - Xin Hao
- grid.452743.30000 0004 1788 4869Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, 225009 Yangzhou, Jiangsu China ,grid.268415.cDepartment of Clinical Medicine, Medical College, Yangzhou University, 225001 Yangzhou, Jiangsu China
| | - Jiajia Li
- grid.452743.30000 0004 1788 4869Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, 225009 Yangzhou, Jiangsu China
| | - Siwei Weng
- grid.268415.cDepartment of stomatology, Clinical Traditional Chinese Medicine College of Yangzhou University, 225000 Yangzhou, Jiangsu China
| | - Jiaxin Wang
- grid.268415.cDepartment of Clinical Medicine, Medical College, Yangzhou University, 225001 Yangzhou, Jiangsu China
| | - Tiantian Zhao
- grid.268415.cDepartment of Clinical Medicine, Medical College, Yangzhou University, 225001 Yangzhou, Jiangsu China
| | - Wenqian Li
- grid.268415.cDepartment of Clinical Medicine, Medical College, Yangzhou University, 225001 Yangzhou, Jiangsu China
| | - Xiaolin Hu
- grid.268415.cDepartment of Clinical Medicine, Medical College, Yangzhou University, 225001 Yangzhou, Jiangsu China
| | - Bing Deng
- grid.452743.30000 0004 1788 4869Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, 225009 Yangzhou, Jiangsu China
| | - Jianguo Gu
- grid.412755.00000 0001 2166 7427Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558 Japan
| | - Qinglei Hang
- grid.412755.00000 0001 2166 7427Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558 Japan ,grid.240145.60000 0001 2291 4776Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| |
Collapse
|
7
|
Vermeulen S, Birgani ZT, Habibovic P. Biomaterial-induced pathway modulation for bone regeneration. Biomaterials 2022; 283:121431. [DOI: 10.1016/j.biomaterials.2022.121431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/28/2022] [Accepted: 02/17/2022] [Indexed: 12/18/2022]
|
8
|
Rapraeger AC. Syndecans and Their Synstatins: Targeting an Organizer of Receptor Tyrosine Kinase Signaling at the Cell-Matrix Interface. Front Oncol 2021; 11:775349. [PMID: 34778093 PMCID: PMC8578902 DOI: 10.3389/fonc.2021.775349] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 09/27/2021] [Indexed: 01/11/2023] Open
Abstract
Receptor tyrosine kinases (RTKs) and integrin matrix receptors have well-established roles in tumor cell proliferation, invasion and survival, often functioning in a coordinated fashion at sites of cell-matrix adhesion. Central to this coordination are syndecans, another class of matrix receptor, that organize RTKs and integrins into functional units, relying on docking motifs in the syndecan extracellular domains to capture and localize RTKs (e.g., EGFR, IGF-1R, VEGFR2, HER2) and integrins (e.g., αvβ3, αvβ5, α4β1, α3β1, α6β4) to sites of adhesion. Peptide mimetics of the docking motifs in the syndecans, called “synstatins”, prevent assembly of these receptor complexes, block their signaling activities and are highly effective against tumor cell invasion and survival and angiogenesis. This review describes our current understanding of these four syndecan-coupled mechanisms and their inhibitory synstatins (SSTNIGF1R, SSTNVEGFR2, SSTNVLA-4, SSTNEGFR and SSTNHER2).
Collapse
Affiliation(s)
- Alan C Rapraeger
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
9
|
Li L, Chen H, He S, Sa G. The adhesive heterogeneity of different compartments of oral mucosal rete ridges. Exp Dermatol 2021; 31:413-419. [PMID: 34665894 DOI: 10.1111/exd.14471] [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: 05/17/2021] [Revised: 09/08/2021] [Accepted: 10/17/2021] [Indexed: 01/18/2023]
Abstract
Rete ridges play a critical role in maintaining epidermal structure and mechanical properties. Notably, rete ridges can be divided into three compartments: the base, slope and tip. The present study aims to explore whether these three compartments have distinct adhesive functions. We collected 28 normal masticatory mucosae to prepare paraffin-embedded sections. Immunohistochemistry and immunofluorescent staining were used to analyse the expression pattern of integrin α6 and β4 in different compartments of the rete ridges. To observe whether the different compartments had distinct adhesive forces, dermal-epidermal junction separation experiments were performed by peeling the oral epithelium from the lamina propria after treatment with cold saline for 72 h. The results showed that integrin α6 and β4 prefer the basal layer keratinocytes closely adjacent to the base compartment of the rete ridges. The oral mucosal epithelium separated from the underlying lamina propria at the tip of rete ridges when they were peeled after the cold saline treatment. In conclusion, the adhesive force of the basal layer keratinocytes at the base of the rete ridges is stronger than at the tip.
Collapse
Affiliation(s)
- Lin Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Heng Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Sangang He
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guoliang Sa
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Te Molder L, de Pereda JM, Sonnenberg A. Regulation of hemidesmosome dynamics and cell signaling by integrin α6β4. J Cell Sci 2021; 134:272177. [PMID: 34523678 DOI: 10.1242/jcs.259004] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Hemidesmosomes (HDs) are specialized multiprotein complexes that connect the keratin cytoskeleton of epithelial cells to the extracellular matrix (ECM). In the skin, these complexes provide stable adhesion of basal keratinocytes to the underlying basement membrane. Integrin α6β4 is a receptor for laminins and plays a vital role in mediating cell adhesion by initiating the assembly of HDs. In addition, α6β4 has been implicated in signal transduction events that regulate diverse cellular processes, including proliferation and survival. In this Review, we detail the role of α6β4 in HD assembly and beyond, and we discuss the molecular mechanisms that regulate its function.
Collapse
Affiliation(s)
- Lisa Te Molder
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Jose M de Pereda
- Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Salamanca, 37007 Salamanca, Spain
| | - Arnoud Sonnenberg
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
11
|
Yang H, Xu Z, Peng Y, Wang J, Xiang Y. Integrin β4 as a Potential Diagnostic and Therapeutic Tumor Marker. Biomolecules 2021; 11:biom11081197. [PMID: 34439865 PMCID: PMC8394641 DOI: 10.3390/biom11081197] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/09/2021] [Accepted: 08/11/2021] [Indexed: 11/05/2022] Open
Abstract
Integrin β4 (ITGβ4) is a class of transmembrane adhesion molecules composed of hemidesmosomes (HDs). Its unique long intracellular domain provides intricate signal transduction functions. These signal transduction effects are especially prominent in tumors. Many recent studies have shown that integrin β4 is differentially expressed in various tumors, and it plays a vital role in tumor invasion, proliferation, epithelial–mesenchymal transition, and angiogenesis. Therefore, we categorize the research related to integrin β4, starting from its structure and function in tumor tissues, and provide a basic description. Based on its structure and function, we believe that integrin β4 can be used as a tumor marker. In clinical practice, it is described as a diagnostic marker for the targeted treatment of cancer and will be helpful in the clinical diagnosis and treatment of tumors.
Collapse
Affiliation(s)
- Haoyu Yang
- School of Basic Medical Science, Central South University, Changsha 410013, China; (H.Y.); (Z.X.); (Y.P.)
| | - Zixuan Xu
- School of Basic Medical Science, Central South University, Changsha 410013, China; (H.Y.); (Z.X.); (Y.P.)
| | - Yuqian Peng
- School of Basic Medical Science, Central South University, Changsha 410013, China; (H.Y.); (Z.X.); (Y.P.)
| | - Jiali Wang
- Xiang Ya School of Medicine, Central South University, Changsha 410013, China;
| | - Yang Xiang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410013, China
- Correspondence: ; Tel.:+86-139-7312-8943
| |
Collapse
|
12
|
Beauséjour M, Boutin A, Vachon PH. Anoikis and the Human Gut Epithelium in Health and Disease. ANOIKIS 2021:95-126. [DOI: 10.1007/978-3-030-73856-3_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
13
|
Zemljič Jokhadar Š, Stojković B, Vidak M, Sorčan T, Liovic M, Gouveia M, Travasso RDM, Derganc J. Cortical stiffness of keratinocytes measured by lateral indentation with optical tweezers. PLoS One 2021; 15:e0231606. [PMID: 33382707 PMCID: PMC7774922 DOI: 10.1371/journal.pone.0231606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 12/04/2020] [Indexed: 01/20/2023] Open
Abstract
Keratin intermediate filaments are the principal structural element of epithelial cells. Their importance in providing bulk cellular stiffness is well recognized, but their role in the mechanics of cell cortex is less understood. In this study, we therefore compared the cortical stiffness of three keratinocyte lines: primary wild type cells (NHEK2), immortalized wild type cells (NEB1) and immortalized mutant cells (KEB7). The cortical stiffness was measured by lateral indentation of cells with AOD-steered optical tweezers without employing any moving mechanical elements. The method was validated on fixed cells and Cytochalasin-D treated cells to ensure that the observed variations in stiffness within a single cell line were not a consequence of low measurement precision. The measurements of the cortical stiffness showed that primary wild type cells were significantly stiffer than immortalized wild type cells, which was also detected in previous studies of bulk elasticity. In addition, a small difference between the mutant and the wild type cells was detected, showing that mutation of keratin impacts also the cell cortex. Thus, our results indicate that the role of keratins in cortical stiffness is not negligible and call for further investigation of the mechanical interactions between keratins and elements of the cell cortex.
Collapse
Affiliation(s)
- Špela Zemljič Jokhadar
- Institute for Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Biljana Stojković
- Institute for Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Marko Vidak
- Medical Center for Molecular Biology, Institute for Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tjaša Sorčan
- Medical Center for Molecular Biology, Institute for Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mirjana Liovic
- Medical Center for Molecular Biology, Institute for Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Marcos Gouveia
- Department of Physics, Centro de Física da Universidade de Coimbra (CFisUC), University of Coimbra, Coimbra, Portugal
| | - Rui D. M. Travasso
- Department of Physics, Centro de Física da Universidade de Coimbra (CFisUC), University of Coimbra, Coimbra, Portugal
| | - Jure Derganc
- Institute for Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- * E-mail:
| |
Collapse
|
14
|
Song G, Luo BH. Atypical structure and function of integrin α V β 8. J Cell Physiol 2020; 236:4874-4887. [PMID: 33368230 DOI: 10.1002/jcp.30242] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/10/2020] [Accepted: 12/12/2020] [Indexed: 12/12/2022]
Abstract
Integrins are heterodimeric transmembrane proteins that play important roles in various biological processes. Most integrins serve as adhesion molecules and transmit bidirectional signaling across the cell membrane through global conformational changes from the bent closed to the extended open conformation. However, integrin β8 is distinctive in structure and function. Its cytoplasmic domain lacks the conserved protein-binding sequence, which is important in transmitting inside-out signals, suggesting that integrin β8 may have a different activation mechanism or lack such signaling. In addition, the ligand-binding or activating metal ion Mn2+ does not induce a global conformational change in integrin β8 . It may have only one conformation, that is, an extended, closed conformation, but with high affinity for ligands under physiological conditions, and is, therefore, considered an atypical integrin member. The extended structure and high ligand-binding affinity of integrin αv β8 make it ideal for encountering and binding ligands expressed on an opposing cell or in the extracellular matrix. In this review, we summarize the progress in integrin β8 research with a focus on its distinctive function and structure among integrin members.
Collapse
Affiliation(s)
- Guannan Song
- Department of Life Science, University of Louisiana State University, Baton Rouge, Louisiana, USA
| | - Bing-Hao Luo
- Department of Life Science, University of Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|
15
|
Tong Z, Liu Y, Xia R, Chang Y, Hu Y, Liu P, Zhai Z, Zhang J, Li H. F-actin Regulates Osteoblastic Differentiation of Mesenchymal Stem Cells on TiO 2 Nanotubes Through MKL1 and YAP/TAZ. NANOSCALE RESEARCH LETTERS 2020; 15:183. [PMID: 32965618 PMCID: PMC7511505 DOI: 10.1186/s11671-020-03415-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 09/14/2020] [Indexed: 05/02/2023]
Abstract
Titanium and titanium alloys are widely used in orthopedic implants. Modifying the nanotopography provides a new strategy to improve osseointegration of titanium substrates. Filamentous actin (F-actin) polymerization, as a mechanical loading structure, is generally considered to be involved in cell migration, endocytosis, cell division, and cell shape maintenance. Whether F-actin is involved and how it functions in nanotube-induced osteogenic differentiation of mesenchymal stem cells (MSCs) remain to be elucidated. In this study, we fabricated TiO2 nanotubes on the surface of a titanium substrate by anodic oxidation and characterized their features by scanning electron microscopy (SEM), X-ray energy dispersive analysis (EDS), and atomic force microscopy (AFM). Alkaline phosphatase (ALP) staining, Western blotting, qRT-PCR, and immunofluorescence staining were performed to explore the osteogenic potential, the level of F-actin, and the expression of MKL1 and YAP/TAZ. Our results showed that the inner diameter and roughness of TiO2 nanotubes increased with the increase of the anodic oxidation voltage from 30 to 70 V, while their height was 2 μm consistently. Further, the larger the tube diameter, the stronger the ability of TiO2 nanotubes to promote osteogenic differentiation of MSCs. Inhibiting F-actin polymerization by Cyto D inhibited osteogenic differentiation of MSCs as well as the expression of proteins contained in focal adhesion complexes such as vinculin (VCL) and focal adhesion kinase (FAK). In contrast, after Jasp treatment, polymerization of F-actin enhanced the expression of RhoA and transcription factors YAP/TAZ. Based on these data, we concluded that TiO2 nanotubes facilitated the osteogenic differentiation of MSCs, and this ability was enhanced with the increasing diameter of the nanotubes within a certain range (30-70 V). F-actin mediated this process through MKL1 and YAP/TAZ.
Collapse
Affiliation(s)
- Zhicheng Tong
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Yanchang Liu
- Department of Orthopedics, The Second Hospital of Anhui Medical University, Hefei, 230601, Anhui, People's Republic of China
| | - Runzhi Xia
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Yongyun Chang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Yi Hu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Pengcheng Liu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Zanjing Zhai
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Jingwei Zhang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Huiwu Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
16
|
Wu M, Yang Y, Yuan L, Yang M, Wang L, Du X, Qin L, Wu S, Xiang Y, Qu X, Liu H, Qin X, Liu C. DNA methylation down-regulates integrin β4 expression in asthmatic airway epithelial cells. Clin Exp Allergy 2020; 50:1127-1139. [PMID: 32618381 DOI: 10.1111/cea.13697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/10/2020] [Accepted: 06/17/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Integrin β4 (ITGB4) is a hemi-desmosome protein which is downregulated in the airway epithelial cells of asthma patients. The proximal promoters and exons of ITGB4 contain CpG islands or multiple CpG sites both in human and mice, which indicated the possible methylation regulation of ITGB4 in airway epithelial cells. OBJECTIVE We sought to unveil that DNA methylation regulates the decreased ITGB4 during the pathogenesis of asthma. METHODS Mice were exposed to house dust mite (HDM) extracts to construct an asthma model. 5-Aza-2'-deoxycytidine (5-AZA) or dexamethasone (DEX) were added in the last two weeks. Besides, the primary human bronchial epithelial (HBE) cells were incubated for the detection of ITGB4 expression and methylation status after HDM stress. Furthermore, DNA methylation of ITGB4 in peripheral blood was measured in asthma patients. Logistic regression was employed to evaluate the association between methylation sites and asthma patients' ages in the control of potential confounders. Moreover, the correlations between differentially methylated sites (DMSs) and clinical parameters in asthma patients were assessed. Finally, the ability of candidate DMSs to predict asthma was evaluated by receiver operating characteristic (ROC) analysis and principal component analysis (PCA). RESULTS We found that in HDM-stressed asthma model, DNA methylation regulated the reduced ITGB4 expression in airway epithelial cells. Moreover, alteration in the specific CpG sites (chr17:73717720 and chr17:73717636) of ITGB4 may regulate ITGB4 expression and further may be associated with the clinically phenotypic of asthma. The specific DMSs of ITGB4 in peripheral blood can distinguish asthma patients from healthy controls (HCs) effectively. CONCLUSIONS AND CLINICAL RELEVANCE This study confirmed that DNA methylation regulates the decreased expression of ITGB4 in the airway epithelial cells of asthma patients. These results supply some useful insights to the mechanism of the decreased ITGB4 in asthmatic airway epithelial and provide possible targets for early prediction and screening of asthma.
Collapse
Affiliation(s)
- Mengping Wu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Yu Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Lin Yuan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Ming Yang
- Faculty of Health and Medicine, Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW, Australia
| | - Leyuan Wang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xizi Du
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Ling Qin
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Shuangyan Wu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Chi Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China.,Research Center of China-Africa Infectious Diseases, Xiangya School of Medicine Central South University, Changsha, China
| |
Collapse
|
17
|
Gouveia M, Zemljič-Jokhadar Š, Vidak M, Stojkovič B, Derganc J, Travasso R, Liovic M. Keratin Dynamics and Spatial Distribution in Wild-Type and K14 R125P Mutant Cells-A Computational Model. Int J Mol Sci 2020; 21:E2596. [PMID: 32283594 PMCID: PMC7177522 DOI: 10.3390/ijms21072596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 12/30/2022] Open
Abstract
Keratins are one of the most abundant proteins in epithelial cells. They form a cytoskeletal filament network whose structural organization seriously conditions its function. Dynamic keratin particles and aggregates are often observed at the periphery of mutant keratinocytes related to the hereditary skin disorder epidermolysis bullosa simplex, which is due to mutations in keratins 5 and 14. To account for their emergence in mutant cells, we extended an existing mathematical model of keratin turnover in wild-type cells and developed a novel 2D phase-field model to predict the keratin distribution inside the cell. This model includes the turnover between soluble, particulate and filamentous keratin forms. We assumed that the mutation causes a slowdown in the assembly of an intermediate keratin phase into filaments, and demonstrated that this change is enough to account for the loss of keratin filaments in the cell's interior and the emergence of keratin particles at its periphery. The developed mathematical model is also particularly tailored to model the spatial distribution of keratins as the cell changes its shape.
Collapse
Affiliation(s)
- Marcos Gouveia
- CFisUC, Center for Physics of the University of Coimbra, Department of Physics, University of Coimbra, R Larga, 3004-516 Coimbra, Portugal
| | - Špela Zemljič-Jokhadar
- Institute for Biophysics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (Š.Z.-J.); (B.S.); (J.D.)
| | - Marko Vidak
- Medical Center for Molecular Biology, Institute for Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (M.V.); (M.L.)
| | - Biljana Stojkovič
- Institute for Biophysics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (Š.Z.-J.); (B.S.); (J.D.)
| | - Jure Derganc
- Institute for Biophysics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (Š.Z.-J.); (B.S.); (J.D.)
| | - Rui Travasso
- CFisUC, Center for Physics of the University of Coimbra, Department of Physics, University of Coimbra, R Larga, 3004-516 Coimbra, Portugal
| | - Mirjana Liovic
- Medical Center for Molecular Biology, Institute for Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (M.V.); (M.L.)
| |
Collapse
|
18
|
Manso JA, Gómez-Hernández M, Carabias A, Alonso-García N, García-Rubio I, Kreft M, Sonnenberg A, de Pereda JM. Integrin α6β4 Recognition of a Linear Motif of Bullous Pemphigoid Antigen BP230 Controls Its Recruitment to Hemidesmosomes. Structure 2019; 27:952-964.e6. [PMID: 31006587 DOI: 10.1016/j.str.2019.03.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/13/2019] [Accepted: 03/22/2019] [Indexed: 11/25/2022]
Abstract
Mechanical stability of epithelia requires firm attachment to the basement membrane via hemidesmosomes. Dysfunction of hemidesmosomal proteins causes severe skin-blistering diseases. Two plakins, plectin and BP230 (BPAG1e), link the integrin α6β4 to intermediate filaments in epidermal hemidesmosomes. Here, we show that a linear sequence within the isoform-specific N-terminal region of BP230 binds to the third and fourth FnIII domains of β4. The crystal structure of the complex and mutagenesis analysis revealed that BP230 binds between the two domains of β4. BP230 induces closing of the two FnIII domains that are locked in place by an interdomain ionic clasp required for binding. Disruption of BP230-β4 binding prevents recruitment of BP230 to hemidesmosomes in human keratinocytes, revealing a key role of this interaction for hemidesmosome assembly. Phosphomimetic substitutions in β4 and BP230 destabilize the complex. Thus, our study provides insights into the architecture of hemidesmosomes and potential mechanisms of regulation.
Collapse
Affiliation(s)
- José A Manso
- Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas - University of Salamanca, Campus Unamuno, 37007 Salamanca, Spain
| | - María Gómez-Hernández
- Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas - University of Salamanca, Campus Unamuno, 37007 Salamanca, Spain
| | - Arturo Carabias
- Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas - University of Salamanca, Campus Unamuno, 37007 Salamanca, Spain
| | - Noelia Alonso-García
- Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas - University of Salamanca, Campus Unamuno, 37007 Salamanca, Spain
| | - Inés García-Rubio
- Centro Universitario de la Defensa, Ctra. Huesca s/n, 50090 Zaragoza, Spain
| | - Maaike Kreft
- Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Arnoud Sonnenberg
- Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - José M de Pereda
- Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas - University of Salamanca, Campus Unamuno, 37007 Salamanca, Spain.
| |
Collapse
|
19
|
A novel miR-365-3p/EHF/keratin 16 axis promotes oral squamous cell carcinoma metastasis, cancer stemness and drug resistance via enhancing β5-integrin/c-met signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:89. [PMID: 30782177 PMCID: PMC6381632 DOI: 10.1186/s13046-019-1091-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 02/06/2019] [Indexed: 01/17/2023]
Abstract
Background Targeting the c-Met signaling pathway has become a therapeutic strategy in multiple types of cancer. We unveiled a novel c-Met regulating mechanism that could be applied as a modality for oral squamous cell carcinoma (OSCC) therapy. Methods Upregulation of keratin 16 (KRT16) was found by comparing isogenic pairs of low and high invasive human OSCC lines via microarray analysis. OSCC cells with ectopic expression or silencing of KRT16 were used to scrutinize functional roles and associated molecular mechanisms. Results We observed that high KRT16 expression significantly correlated with poorer pathological differentiation, advanced stages, increased lymph nodes metastasis, and decreased survival rate from several Taiwanese OSCC patient cohorts. We further revealed that miR-365-3p could target ETS homologous factor (EHF), a KRT16 transcription factor, to decrease migration, invasion, metastasis and chemoresistance in OSCC cells via inhibition of KRT16. Under confocal microscopic examination, c-Met was found possibly partially associates with KRT16 through β5-integrin. Colocalization of these three proteins may facilitate c-Met and β5-integrin–mediated signaling in OSCC cells. Depletion of KRT16 led to increased protein degradation of β5-integrin and c-Met through a lysosomal pathway leading to inhibition of their downstream Src/STAT3/FAK/ERK signaling in OSCC cells. Knockdown of KRT16 enhanced chemosensitivity of OSCC towards 5-fluorouracil (5-FU). Various combination of c-Met inhibitor (foretinib), protein tyrosine kinase inhibitor (genistein), β5-integrin antibody, and 5-FU markedly augmented cytotoxic effects in OSCC cells as well as tumor killing effects in vitro and in vivo. Conclusions Our data indicate that targeting a novel miR-365-3p/EHF/KRT16/β5-integrin/c-Met signaling pathway could improve treatment efficacy in OSCC. Electronic supplementary material The online version of this article (10.1186/s13046-019-1091-5) contains supplementary material, which is available to authorized users.
Collapse
|
20
|
Effects of quercetin on human oral keratinocytes during re-epithelialization: An in vitro study. Arch Oral Biol 2018; 95:187-194. [DOI: 10.1016/j.archoralbio.2018.08.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 08/09/2018] [Accepted: 08/10/2018] [Indexed: 01/06/2023]
|
21
|
Beauséjour M, Boutin A, Vachon PH. Anoikis Regulation: Complexities, Distinctions, and Cell Differentiation. APOPTOSIS AND BEYOND 2018:145-182. [DOI: 10.1002/9781119432463.ch8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
22
|
Fujiwara S, Matsui TS, Ohashi K, Deguchi S, Mizuno K. Solo, a RhoA-targeting guanine nucleotide exchange factor, is critical for hemidesmosome formation and acinar development in epithelial cells. PLoS One 2018; 13:e0195124. [PMID: 29672603 PMCID: PMC5909619 DOI: 10.1371/journal.pone.0195124] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/17/2018] [Indexed: 11/18/2022] Open
Abstract
Cell-substrate adhesions are essential for various physiological processes, including embryonic development and maintenance of organ functions. Hemidesmosomes (HDs) are multiprotein complexes that attach epithelial cells to the basement membrane. Formation and remodeling of HDs are dependent on the surrounding mechanical environment; however, the upstream signaling mechanisms are not well understood. We recently reported that Solo (also known as ARHGEF40), a guanine nucleotide exchange factor targeting RhoA, binds to keratin8/18 (K8/K18) intermediate filaments, and that their interaction is important for force-induced actin and keratin cytoskeletal reorganization. In this study, we show that Solo co-precipitates with an HD protein, β4-integrin. Co-precipitation assays revealed that the central region (amino acids 330–1057) of Solo binds to the C-terminal region (1451–1752) of β4-integrin. Knockdown of Solo significantly suppressed HD formation in MCF10A mammary epithelial cells. Similarly, knockdown of K18 or treatment with Y-27632, a specific inhibitor of Rho-associated kinase (ROCK), suppressed HD formation. As Solo knockdown or Y-27632 treatment is known to disorganize K8/K18 filaments, these results suggest that Solo is involved in HD formation by regulating K8/K18 filament organization via the RhoA-ROCK signaling pathway. We also showed that knockdown of Solo impairs acinar formation in MCF10A cells cultured in 3D Matrigel. In addition, Solo accumulated at the site of traction force generation in 2D-cultured MCF10A cells. Taken together, these results suggest that Solo plays a crucial role in HD formation and acinar development in epithelial cells by regulating mechanical force-induced RhoA activation and keratin filament organization.
Collapse
Affiliation(s)
- Sachiko Fujiwara
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka, Japan
- Research Fellow of the Japanese Society for the Promotion of Science, Kojimachi, Chiyoda-ku, Tokyo, Japan
- * E-mail: (SF); (SD); (KM)
| | - Tsubasa S. Matsui
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka, Japan
| | - Kazumasa Ohashi
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Shinji Deguchi
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka, Japan
- * E-mail: (SF); (SD); (KM)
| | - Kensaku Mizuno
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
- * E-mail: (SF); (SD); (KM)
| |
Collapse
|
23
|
Kariya Y, Oyama M, Hashimoto Y, Gu J, Kariya Y. β4-Integrin/PI3K Signaling Promotes Tumor Progression through the Galectin-3- N-Glycan Complex. Mol Cancer Res 2018; 16:1024-1034. [PMID: 29549127 DOI: 10.1158/1541-7786.mcr-17-0365] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 02/07/2018] [Accepted: 03/12/2018] [Indexed: 11/16/2022]
Abstract
Malignant transformation is associated with aberrant N-glycosylation, but the role of protein N-glycosylation in cancer progression remains poorly defined. β4-integrin is a major carrier of N-glycans and is associated with poor prognosis, tumorigenesis, and metastasis. Here, N-glycosylation of β4-integrin contributes to the activation of signaling pathways that promote β4-dependent tumor development and progression. Increased expression of β1,6GlcNAc-branched N-glycans was found to be colocalized with β4-integrin in human cutaneous squamous cell carcinoma tissues, and that the β1,6GlcNAc residue was abundant on β4-integrin in transformed keratinocytes. Interruption of β1,6GlcNAc-branching formation on β4-integrin with the introduction of bisecting GlcNAc by N-acetylglucosaminyltransferase III overexpression was correlated with suppression of cancer cell migration and tumorigenesis. N-Glycan deletion on β4-integrin impaired β4-dependent cancer cell migration, invasion, and growth in vitro and diminished tumorigenesis and proliferation in vivo The reduced abilities of β4-integrin were accompanied with decreased phosphoinositol-3 kinase (PI3K)/Akt signals and were restored by the overexpression of the constitutively active p110 PI3K subunit. Binding of galectin-3 to β4-integrin via β1,6GlcNAc-branched N-glycans promoted β4-integrin-mediated cancer cell adhesion and migration. In contrast, a neutralizing antibody against galectin-3 attenuated β4-integrin N-glycan-mediated PI3K activation and inhibited the ability of β4-integrin to promote cell motility. Furthermore, galectin-3 knockdown by shRNA suppressed β4-integrin N-glycan-mediated tumorigenesis. These findings provide a novel role for N-glycosylation of β4-integrin in tumor development and progression, and the regulatory mechanism for β4-integrin/PI3K signaling via the galectin-3-N-glycan complex.Implications:N-Glycosylation of β4-integrin plays a functional role in promoting tumor development and progression through PI3K activation via the galectin-3-N-glycan complex. Mol Cancer Res; 16(6); 1024-34. ©2018 AACR.
Collapse
Affiliation(s)
- Yukiko Kariya
- Department of Biochemistry, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Midori Oyama
- Department of Biochemistry, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yasuhiro Hashimoto
- Department of Biochemistry, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Miyagi, Japan
| | - Yoshinobu Kariya
- Department of Biochemistry, Fukushima Medical University School of Medicine, Fukushima, Japan.
| |
Collapse
|
24
|
García-Mariscal A, Li H, Pedersen E, Peyrollier K, Ryan KM, Stanley A, Quondamatteo F, Brakebusch C. Loss of RhoA promotes skin tumor formation and invasion by upregulation of RhoB. Oncogene 2018; 37:847-860. [PMID: 29059167 DOI: 10.1038/onc.2017.333] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 07/26/2017] [Accepted: 08/12/2017] [Indexed: 02/06/2023]
Abstract
Cellular movement is controlled by small GTPases, such as RhoA. Although migration is crucial for cancer cell invasion, the specific role of RhoA in tumor formation is unclear. Inducing skin tumors in mice with a keratinocyte-restricted loss of RhoA, we observed increased tumor frequency, growth and invasion. In vitro invasion assays revealed that in the absence of RhoA cell invasiveness is increased in a Rho-associated protein kinase (ROCK) activation and cell contraction-dependent manner. Surprisingly, loss of RhoA causes increased Rho signaling via overcompensation by RhoB because of reduced lysosomal degradation of RhoB in Gamma-aminobutyric acid receptor-associated protein (GABARAP)+ autophagosomes and endosomes. In the absence of RhoA, RhoB relocalized to the plasma membrane and functionally replaced RhoA with respect to invasion, clonogenic growth and survival. Our data demonstrate for the first time that RhoA is a tumor suppressor in 7,12-dimethylbenz[a]anthracene/12-O-tetradecanoylphorbol 13-acetate skin carcinogenesis and identify Rho signaling dependent on RhoA and RhoB as a potent driver of tumor progression.
Collapse
Affiliation(s)
- A García-Mariscal
- Biotech Research and Innovation Centre, BRIC, University of Copenhagen, Copenhagen, Denmark
| | - H Li
- Biotech Research and Innovation Centre, BRIC, University of Copenhagen, Copenhagen, Denmark
| | - E Pedersen
- Biotech Research and Innovation Centre, BRIC, University of Copenhagen, Copenhagen, Denmark
| | - K Peyrollier
- Biotech Research and Innovation Centre, BRIC, University of Copenhagen, Copenhagen, Denmark
| | | | - A Stanley
- Skin and Extracellular Matrix Research Group, Anatomy, NUI, Galway, Ireland
| | - F Quondamatteo
- Skin and Extracellular Matrix Research Group, Anatomy, NUI, Galway, Ireland
| | - C Brakebusch
- Biotech Research and Innovation Centre, BRIC, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
Chen WW, Tjin MS, Chua AWC, Lee ST, Tay CY, Fong E. Probing the Role of Integrins in Keratinocyte Migration Using Bioengineered Extracellular Matrix Mimics. ACS APPLIED MATERIALS & INTERFACES 2017; 9:36483-36492. [PMID: 28967740 DOI: 10.1021/acsami.7b06959] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Bioengineered extracellular matrix (ECM) mimetic materials have tunable properties and can be engineered to elicit desirable cellular responses for wound repair and tissue regeneration. By incorporating relevant cell-instructive domains, bioengineered ECM mimics can be designed to provide well-defined ECM-specific cues to influence cell motility and differentiation. More importantly, bioengineered ECM surfaces are ideal platforms for studying cell-material interactions without the need to genetically alter the cells. Here, we showed that bioengineered ECM mimics can be employed to clarify the role of integrins in keratinocyte migration. Particularly, the roles of α5β1 and α3β1 in keratinocytes were examined, given their known importance in keratinocyte motility. Two recombinant proteins were constructed; each protein contains a functional domain taken from fibronectin (FN-mimic) and laminin-332 (LN-mimic), designed to bind α5β1 and α3β1, respectively. We examined how patient-derived primary human keratinocytes migrate when sparsely seeded as well as when allowed to move collectively. We found, consistently, that FN-mimic promoted cell migration while the LN-mimic did not support cell motility. We showed that, when keratinocytes utilize α5β1 integrins on FN-mimics, they were able to form stable focal adhesion plaques and stabilized lamellipodia. On the other hand, keratinocytes on LN-mimic utilized primarily α3β1 integrins for migration and, strikingly, cells were unable to activate Rac1 and form stable focal adhesion plaques. Taken together, employment of our bioengineered mimics has allowed us to clarify the roles of α5β1 and α3β1 integrins in keratinocyte migration, as well as further provided a mechanistic explanation for their differences.
Collapse
Affiliation(s)
- Wilhelm W Chen
- School of Materials Science and Engineering, Nanyang Technological University , N4.1, 50 Nanyang Avenue, Singapore 639798, Singapore
- Mechanobiology Institute, National University of Singapore , 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Monica S Tjin
- Program in Cardiovascular and Metabolic Disorder, Duke-NUS Medical School , 8 College Road, Singapore 169857, Singapore
| | - Alvin W C Chua
- Department of Plastic Reconstructive & Aesthetic Surgery, Singapore General Hospital , Outram Road, Singapore 169608, Singapore
| | - Seng Teik Lee
- Department of Plastic Reconstructive & Aesthetic Surgery, Singapore General Hospital , Outram Road, Singapore 169608, Singapore
| | - Chor Yong Tay
- School of Materials Science and Engineering, Nanyang Technological University , N4.1, 50 Nanyang Avenue, Singapore 639798, Singapore
- School of Biological Sciences, Nanyang Technological University , 60 Nanyang Drive, Singapore 637551, Singapore
| | - Eileen Fong
- School of Materials Science and Engineering, Nanyang Technological University , N4.1, 50 Nanyang Avenue, Singapore 639798, Singapore
| |
Collapse
|
26
|
Kariya Y, Kariya Y, Gu J. Roles of Integrin α6β4 Glycosylation in Cancer. Cancers (Basel) 2017; 9:cancers9070079. [PMID: 28678156 PMCID: PMC5532615 DOI: 10.3390/cancers9070079] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 06/30/2017] [Accepted: 06/30/2017] [Indexed: 01/15/2023] Open
Abstract
Malignant transformation is accompanied with aberrant glycosylation of proteins. Such changes in glycan structure also occur in the integrins, which are a large family of cell surface receptors for the extracellular matrix and play key roles in tumor progression. There is now increasing evidence that glycosylation of integrins affects cellular signaling and interaction with the extracellular matrix, receptor tyrosine kinases, and galectins, thereby regulating cell adhesion, motility, growth, and survival. Integrin α6β4 is a receptor for laminin-332 and the increased expression level is correlated with malignant progression and poor survival in various types of cancers. Recent studies have revealed that integrin α6β4 plays central roles in tumorigenesis and the metastatic process. In this review, we summarize our current understanding of the molecular mechanisms of tumor progression driven by integrin α6β4 and also discuss the modification of glycans on integrin β4 subunit to address the important roles of glycan in integrin-mediated tumor progression.
Collapse
Affiliation(s)
- Yoshinobu Kariya
- Department of Biochemistry, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima City, Fukushima 960-1295, Japan.
| | - Yukiko Kariya
- Department of Biochemistry, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima City, Fukushima 960-1295, Japan.
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558, Japan.
| |
Collapse
|
27
|
Takada Y, Takada YK, Fujita M. Crosstalk between insulin-like growth factor (IGF) receptor and integrins through direct integrin binding to IGF1. Cytokine Growth Factor Rev 2017; 34:67-72. [PMID: 28190785 DOI: 10.1016/j.cytogfr.2017.01.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/15/2016] [Accepted: 01/09/2017] [Indexed: 01/25/2023]
Abstract
It has been generally accepted that integrin cell adhesion receptors are involved in growth factor signaling (integrin-growth factor crosstalk), since antagonists to integrins often suppress growth factor signaling. Partly because integrins have been originally identified as cell adhesion receptors to extracellular matrix (ECM) proteins, current models of the crosstalk between IGF1 and integrins propose that ECM ligands (e.g., vitronectin) bind to integrins and IGF1 binds to IGF receptor type 1 (IGF1R), and two separate signals merge inside the cells. Our research proves otherwise. We discovered that IGF1 interacts directly with integrins, and induces integrin-IGF-IGF1R complex formation on the cell surface. IGF1 signaling can be detected in the absence of ECM (anchorage-independent conditions). Integrin antagonists block both ECM-integrin interaction and IGF-integrin interaction, and do not distinguish the two. This is one possible reason why integrin-IGF1 interaction has not been detected. With these new discoveries, we believe that the direct IGF-integrin interaction should be incorporated into models of IGF1 signaling. The integrin-binding defective mutant of IGF1 is defective in inducing IGF signaling, although the mutant still binds to IGF1R. Notably, the IGF1 mutant is dominant-negative and suppresses cell proliferation induced by wt IGF1, and suppresses tumorigenesis in vivo, and thus the IGF1 mutant has potential as a therapeutic.
Collapse
Affiliation(s)
- Yoshikazu Takada
- Departments of Dermatology, Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA 95817, United States; Graduate Institute of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Hsing Street, Taipei 11031, Taiwan, ROC.
| | - Yoko K Takada
- Departments of Dermatology, Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA 95817, United States; Graduate Institute of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Hsing Street, Taipei 11031, Taiwan, ROC
| | - Masaaki Fujita
- Department of Clinical Immunology and Rheumatology, The Tazuke-Kofukai Medical Research Institute, Kitano Hospital, Osaka, Japan
| |
Collapse
|
28
|
Wang M, Nagle RB, Knudsen BS, Rogers GC, Cress AE. A basal cell defect promotes budding of prostatic intraepithelial neoplasia. J Cell Sci 2017; 130:104-110. [PMID: 27609833 PMCID: PMC5394777 DOI: 10.1242/jcs.188177] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 09/02/2016] [Indexed: 12/15/2022] Open
Abstract
Basal cells in a simple secretory epithelium adhere to the extracellular matrix (ECM), providing contextual cues for ordered repopulation of the luminal cell layer. Early high-grade prostatic intraepithelial neoplasia (HG-PIN) tissue has enlarged nuclei and nucleoli, luminal layer expansion and genomic instability. Additional HG-PIN markers include loss of α6β4 integrin or its ligand laminin-332, and budding of tumor clusters into laminin-511-rich stroma. We modeled the invasive budding phenotype by reducing expression of α6β4 integrin in spheroids formed from two normal human stable isogenic prostate epithelial cell lines (RWPE-1 and PrEC 11220). These normal cells continuously spun in culture, forming multicellular spheroids containing an outer laminin-332 layer, basal cells (expressing α6β4 integrin, high-molecular-weight cytokeratin and p63, also known as TP63) and luminal cells that secrete PSA (also known as KLK3). Basal cells were optimally positioned relative to the laminin-332 layer as determined by spindle orientation. β4-integrin-defective spheroids contained a discontinuous laminin-332 layer corresponding to regions of abnormal budding. This 3D model can be readily used to study mechanisms that disrupt laminin-332 continuity, for example, defects in the essential adhesion receptor (β4 integrin), laminin-332 or abnormal luminal expansion during HG-PIN progression.
Collapse
Affiliation(s)
- Mengdie Wang
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA
| | - Raymond B Nagle
- Department of Pathology, College of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA
| | - Beatrice S Knudsen
- Department of Pathology and Laboratory Medicine, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gregory C Rogers
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA
| | - Anne E Cress
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA
| |
Collapse
|
29
|
The opposing roles of laminin-binding integrins in cancer. Matrix Biol 2017; 57-58:213-243. [DOI: 10.1016/j.matbio.2016.08.007] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 08/02/2016] [Accepted: 08/17/2016] [Indexed: 02/06/2023]
|
30
|
Passmore JAS, Williamson AL. Host Immune Responses Associated with Clearance or Persistence of Human Papillomavirus Infections. CURRENT OBSTETRICS AND GYNECOLOGY REPORTS 2016. [DOI: 10.1007/s13669-016-0163-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
31
|
Ranjan A, Gupta P, Srivastava SK. Penfluridol: An Antipsychotic Agent Suppresses Metastatic Tumor Growth in Triple-Negative Breast Cancer by Inhibiting Integrin Signaling Axis. Cancer Res 2016; 76:877-890. [PMID: 26627008 PMCID: PMC4755811 DOI: 10.1158/0008-5472.can-15-1233] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 11/13/2015] [Indexed: 01/01/2023]
Abstract
Metastasis of breast cancer, especially to the brain, is the major cause of mortality. The inability of anticancer agents to cross the blood-brain-barrier represents a critical challenge for successful treatment. In the current study, we investigated the antimetastatic potential of penfluridol, an antipsychotic drug frequently prescribed for schizophrenia with anticancer activity. We show that penfluridol induced apoptosis and reduced the survival of several metastatic triple-negative breast cancer (TNBC) cell lines. In addition, penfluridol treatment significantly reduced the expression of integrin α6, integrin β4, Fak, paxillin, Rac1/2/3, and ROCK1 in vitro. We further evaluated the efficacy of penfluridol in three different in vivo tumor models. We demonstrate that penfluridol administration to an orthotopic model of breast cancer suppressed tumor growth by 49%. On the other hand, penfluridol treatment inhibited the growth of metastatic brain tumors introduced by intracardiac or intracranial injection of breast cancer cells by 90% and 72%, respectively. Penfluridol-treated tumors from all three models exhibited reduced integrin β4 and increased apoptosis. Moreover, chronic administration of penfluridol failed to elicit significant toxic or behavioral side effects in mice. Taken together, our results indicate that penfluridol effectively reduces the growth of primary TNBC tumors and especially metastatic growth in the brain by inhibiting integrin signaling, and prompt further preclinical investigation into repurposing penfluridol for the treatment of metastatic TNBC.
Collapse
Affiliation(s)
- Alok Ranjan
- Department of Biomedical Sciences and Cancer Biology Center, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Parul Gupta
- Department of Biomedical Sciences and Cancer Biology Center, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Sanjay K Srivastava
- Department of Biomedical Sciences and Cancer Biology Center, Texas Tech University Health Sciences Center, Amarillo, Texas.
| |
Collapse
|
32
|
Yang DJ, Lee KS, Ko CM, Moh SH, Song J, Hur LC, Cheon YW, Yang SH, Choi YH, Kim KW. Leucine-enkephalin promotes wound repair through the regulation of hemidesmosome dynamics and matrix metalloprotease. Peptides 2016; 76:57-64. [PMID: 26763532 DOI: 10.1016/j.peptides.2015.12.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 12/03/2015] [Accepted: 12/29/2015] [Indexed: 11/17/2022]
Abstract
The skin responds to environmental stressors by coordinated actions of neuropeptides and their receptors. An endogenous peptide for δ-opioid receptor (DOPr), Leu-enkephalin (L-ENK), is expressed in the skin and its expression is altered in pathological conditions. Although the importance of DOPr is rapidly gaining recognition, the molecular mechanisms underlying its effects on wound healing are largely undefined. We show here that L-ENK induced activation of Erk, P90(RSK), and Elk-1 and promoted the disruption of hemidesmosomes and the expression of matrix metalloprotease (MMP)-2 and MMP-9, important processes for wound healing. Treatment with Erk inhibitor blocked activation of P90(RSK) and Elk-1 and significantly blunted wound repair. Therefore, our results suggest that activation of Erk and its downstream effectors, P90(RSK) and Elk-1, are critical for DOPr-mediated skin homeostasis.
Collapse
Affiliation(s)
- Dong Joo Yang
- Department of Pharmacology, Wonju College of Medicine, Yonsei University, Wonju 26426, Republic of Korea; Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Republic of Korea
| | - Kyung Suk Lee
- Department of Plastic and Reconstructive Surgery, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 52828, Republic of Korea
| | - Chang Mann Ko
- Department of Pharmacology, Wonju College of Medicine, Yonsei University, Wonju 26426, Republic of Korea
| | - Sang Hyun Moh
- Anti-aging Research Institute of BIO-FD&C Co. Ltd., Incheon 21990, Republic of Korea
| | - Jihyeok Song
- Anti-aging Research Institute of BIO-FD&C Co. Ltd., Incheon 21990, Republic of Korea
| | - Lucia C Hur
- Derma-Lucia Skinceuticals LLC, 7500 Escala Drive, Austin, TX 78735, USA
| | - Young Woo Cheon
- Department of Plastic and Reconstructive Surgery, Gachon University Gil Medical Center, Gachon University, School of Medicine, 1198 Guwol-Dong, Namdong-Gu, Incheon 21565, Republic of Korea
| | - Seung Ho Yang
- Department of Pharmacology, Wonju College of Medicine, Yonsei University, Wonju 26426, Republic of Korea
| | - Yun-Hee Choi
- Anti-aging Research Institute of BIO-FD&C Co. Ltd., Incheon 21990, Republic of Korea.
| | - Ki Woo Kim
- Department of Pharmacology, Wonju College of Medicine, Yonsei University, Wonju 26426, Republic of Korea; Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Republic of Korea.
| |
Collapse
|
33
|
Wang H, Jin H, Rapraeger AC. Syndecan-1 and Syndecan-4 Capture Epidermal Growth Factor Receptor Family Members and the α3β1 Integrin Via Binding Sites in Their Ectodomains: NOVEL SYNSTATINS PREVENT KINASE CAPTURE AND INHIBIT α6β4-INTEGRIN-DEPENDENT EPITHELIAL CELL MOTILITY. J Biol Chem 2015; 290:26103-13. [PMID: 26350464 DOI: 10.1074/jbc.m115.679084] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Indexed: 11/06/2022] Open
Abstract
The α6β4 integrin is known to associate with receptor tyrosine kinases when engaged in epithelial wound healing and in carcinoma invasion and survival. Prior work has shown that HER2 associates with α6β4 integrin and syndecan-1 (Sdc1), in which Sdc1 engages the cytoplasmic domain of the β4 integrin subunit allowing HER2-dependent motility and carcinoma cell survival. In contrast, EGFR associates with Sdc4 and the α6β4 integrin, and EGFR-dependent motility depends on cytoplasmic engagement of β4 integrin with Sdc4. However, how HER2 and EGFR assimilate into a complex with the syndecans and integrin, and why kinase capture is syndecan-specific has remained unknown. In the present study, we demonstrate that HER2 is captured via a site, comprised of amino acids 210-240, in the extracellular domain of human Sdc1, and EGFR is captured via an extracellular site comprised of amino acids 87-131 in human Sdc4. Binding assays using purified recombinant proteins demonstrate that the interaction between the EGFR family members and the syndecans is direct. The α3β1 integrin, which is responsible for the motility of the cells, is captured at these sites as well. Peptides based on the interaction motifs in Sdc1 and Sdc4, called synstatins (SSTN210-240 and SSTN87-131) competitively displace the receptor tyrosine kinase and α3β1 integrin from the syndecan with an IC50 of 100-300 nm. The syndecans remain anchored to the α6β4 integrin via its cytoplasmic domain, but the activation of cell motility is disrupted. These novel SSTN peptides are potential therapeutics for carcinomas that depend on these HER2- and EGFR-coupled mechanisms for their invasion and survival.
Collapse
Affiliation(s)
| | | | - Alan C Rapraeger
- From the Department of Human Oncology and the University of Wisconsin Carbone Cancer Center, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, Wisconsin 53705
| |
Collapse
|
34
|
Randhawa V, Acharya V. Integrated network analysis and logistic regression modeling identify stage-specific genes in Oral Squamous Cell Carcinoma. BMC Med Genomics 2015; 8:39. [PMID: 26179909 PMCID: PMC4502639 DOI: 10.1186/s12920-015-0114-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/06/2015] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is associated with substantial mortality and morbidity but, OSCC can be difficult to detect at its earliest stage due to its molecular complexity and clinical behavior. Therefore, identification of key gene signatures at an early stage will be highly helpful. METHODS The aim of this study was to identify key genes associated with progression of OSCC stages. Gene expression profiles were classified into cancer stage-related modules, i.e., groups of genes that are significantly related to a clinical stage. For prioritizing the candidate genes, analysis was further restricted to genes with high connectivity and a significant association with a stage. To assess predictive power of these genes, a classification model was also developed and tested by 5-fold cross validation and on an independent dataset. RESULTS The identified genes were enriched for significant processes and functional pathways, and various genes were found to be directly implicated in OSCC. Forward and stepwise, multivariate logistic regression analyses identified 13 key genes whose expression discriminated early- and late-stage OSCC with predictive accuracy (area under curve; AUC) of ~0.81 in a 5-fold cross-validation strategy. CONCLUSIONS The proposed network-driven integrative analytical approach can identify multiple genes significantly related to an OSCC stage; the classification model that is developed with these genes may help to distinguish cancer stages. The proposed genes and model hold promise for monitoring of OSCC stage progression, and our findings may facilitate cancer detection at an earlier stage, resulting in improved treatment outcomes.
Collapse
Affiliation(s)
- Vinay Randhawa
- Functional Genomics and Complex Systems Laboratory, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Council of Scientific and Industrial Research, Palampur, Himachal Pradesh, India. .,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India.
| | - Vishal Acharya
- Functional Genomics and Complex Systems Laboratory, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Council of Scientific and Industrial Research, Palampur, Himachal Pradesh, India. .,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India.
| |
Collapse
|
35
|
Molecular architecture and function of the hemidesmosome. Cell Tissue Res 2015; 360:529-44. [PMID: 26017636 PMCID: PMC4452579 DOI: 10.1007/s00441-015-2216-6] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 11/03/2014] [Indexed: 01/13/2023]
Abstract
Hemidesmosomes are multiprotein complexes that facilitate the stable adhesion of basal epithelial cells to the underlying basement membrane. The mechanical stability of hemidesmosomes relies on multiple interactions of a few protein components that form a membrane-embedded tightly-ordered complex. The core of this complex is provided by integrin α6β4 and P1a, an isoform of the cytoskeletal linker protein plectin that is specifically associated with hemidesmosomes. Integrin α6β4 binds to the extracellular matrix protein laminin-332, whereas P1a forms a bridge to the cytoplasmic keratin intermediate filament network. Other important components are BPAG1e, the epithelial isoform of bullous pemphigoid antigen 1, BPAG2, a collagen-type transmembrane protein and CD151. Inherited or acquired diseases in which essential components of the hemidesmosome are missing or structurally altered result in tissue fragility and blistering. Modulation of hemidesmosome function is of crucial importance for a variety of biological processes, such as terminal differentiation of basal keratinocytes and keratinocyte migration during wound healing and carcinoma invasion. Here, we review the molecular characteristics of the proteins that make up the hemidesmosome core structure and summarize the current knowledge about how their assembly and turnover are regulated by transcriptional and post-translational mechanisms.
Collapse
|
36
|
Wang Q, Wang Y, Huang X, Liang W, Xiong Z, Xiong Z. Integrin β4 in EMT: an implication of renal diseases. Int J Clin Exp Med 2015; 8:6967-76. [PMID: 26221233 PMCID: PMC4509178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/06/2015] [Indexed: 05/18/2024]
Abstract
Renal fibrosis is a main cause of chronic renal failure. Epithelial-to-mesenchymal transition (EMT) markers play a role in renal fibrosis. Transforming growth factor-β1 (TGF-β1) has been shown to initiate and complete the whole EMT process. It is now well accepted that loss of E-cadherin, EMT marker α-SMA, and connective tissue growth factor (CTGF) expression are key events in the EMT process. We found that by stimulating human renal proximal tubular epithelial (HK-2) cells with TGF-β1, the expression of E-cadherin was down regulated and the expression of α-SMA and CTGF were up regulated in a dose dependent manner. In our present study we also found that integrin β4 and peroxisome proliferators-activated receptor-γ (PPAR-γ) play roles in EMT process, with TGF-β1 stimulation increasing integrin β4 expression in HK2 cells. Integrin β4 and PPARγ were detected in tubulointerstitial tissues, immunohistochemistry analysis showed enhanced expression of integrin β4 in early stage, with over-expression at later stage. In contrast, the expression of PPARγ showed little increased in early stage, but was dramatically decreased at later stage. This is consistent with TGF-β1 inducing EMT. Our immune-precipitation studies show that integrin β4 disassociation with PPARγ is present in E-cadherin signaling. It suggests that PPARγ has a role in EMT inhibition.
Collapse
Affiliation(s)
- Qi Wang
- Department of Nephrology, Peking University Shenzhen Hospital Shenzhen 518036, China
| | - Yan Wang
- Department of Nephrology, Peking University Shenzhen Hospital Shenzhen 518036, China
| | - Xiaoyan Huang
- Department of Nephrology, Peking University Shenzhen Hospital Shenzhen 518036, China
| | - Wei Liang
- Department of Nephrology, Peking University Shenzhen Hospital Shenzhen 518036, China
| | - Zibo Xiong
- Department of Nephrology, Peking University Shenzhen Hospital Shenzhen 518036, China
| | - Zuying Xiong
- Department of Nephrology, Peking University Shenzhen Hospital Shenzhen 518036, China
| |
Collapse
|
37
|
Ganjare A, Bagul N, Kathariya R, Oberoi J. ‘Cell junctions of oral mucosa’- in a nutshell. QSCIENCE CONNECT 2015. [DOI: 10.5339/connect.2015.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Junctional complexes are specialized contacts between neighboring cells and between cells and the extracellular matrix. They play an important role in embryogenesis, growth and development, as well as being the cause of pathologies. These contacts lead to a number of different interactions that have a profound effect on cellular biology. Cell junctions are best visualized using conventional or freeze-fracture electron microscopy, which reveals the interacting plasma membranes are highly specialized in these regions.
Cell adhesion molecules (CAMs) are proteins responsible for homophillic and heterophillic adhesions. They consist of various groups, including cadherins, selectins and intergrins and they facilitate cell adhesion, cell signaling, and motility. Dysregulation of these molecules can lead to various pathologies, for example mucocutaneous diseases and invasion of cancer. This review focuses on the pathophysiology of cell junctions and related diseases.
Collapse
Affiliation(s)
- Anjali Ganjare
- 1Department of Oral Pathology and Microbiology, Dr. D. Y Patil Dental College and Hospital, Dr. D. Y Patil Vidyapeeth, Pune-18, India
| | - Neeta Bagul
- 1Department of Oral Pathology and Microbiology, Dr. D. Y Patil Dental College and Hospital, Dr. D. Y Patil Vidyapeeth, Pune-18, India
| | - Rahul Kathariya
- 2Department of Periodontics and Oral Implantology, Dr. D. Y Patil Dental College and Hospital, Dr. D.Y Patil Vidyapeeth, Pune-18, India
| | - Jyoti Oberoi
- 3Department of Preventive and Pediatric Dentistry, Dr. D. Y Patil School of Dentistry, Nerul, Navi Mumbai- 400706, India
| |
Collapse
|
38
|
Alonso-García N, García-Rubio I, Manso JA, Buey RM, Urien H, Sonnenberg A, Jeschke G, de Pereda JM. Combination of X-ray crystallography, SAXS and DEER to obtain the structure of the FnIII-3,4 domains of integrin α6β4. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2015; 71:969-85. [PMID: 25849406 PMCID: PMC4388270 DOI: 10.1107/s1399004715002485] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 02/05/2015] [Indexed: 03/24/2024]
Abstract
Integrin α6β4 is a major component of hemidesmosomes that mediate the stable anchorage of epithelial cells to the underlying basement membrane. Integrin α6β4 has also been implicated in cell proliferation and migration and in carcinoma progression. The third and fourth fibronectin type III domains (FnIII-3,4) of integrin β4 mediate binding to the hemidesmosomal proteins BPAG1e and BPAG2, and participate in signalling. Here, it is demonstrated that X-ray crystallography, small-angle X-ray scattering and double electron-electron resonance (DEER) complement each other to solve the structure of the FnIII-3,4 region. The crystal structures of the individual FnIII-3 and FnIII-4 domains were solved and the relative arrangement of the FnIII domains was elucidated by combining DEER with site-directed spin labelling. Multiple structures of the interdomain linker were modelled by Monte Carlo methods complying with DEER constraints, and the final structures were selected against experimental scattering data. FnIII-3,4 has a compact and cambered flat structure with an evolutionary conserved surface that is likely to correspond to a protein-interaction site. Finally, this hybrid method is of general application for the study of other macromolecules and complexes.
Collapse
Affiliation(s)
- Noelia Alonso-García
- Instituto de Biología Molecular y Celular del Cancer, Consejo Superior de Investigaciones Científicas – University of Salamanca, Campus Unamuno, 37007 Salamanca, Spain
| | - Inés García-Rubio
- Laboratory of Physical Chemistry, ETH Zürich, Vladimir-Prelog-Weg 2, CH-8093 Zürich, Switzerland
- Centro Universitario de la Defensa, Academia General Militar, Carretera de Huesca s/n, 50090 Zaragoza, Spain
| | - José A. Manso
- Instituto de Biología Molecular y Celular del Cancer, Consejo Superior de Investigaciones Científicas – University of Salamanca, Campus Unamuno, 37007 Salamanca, Spain
| | - Rubén M. Buey
- Instituto de Biología Molecular y Celular del Cancer, Consejo Superior de Investigaciones Científicas – University of Salamanca, Campus Unamuno, 37007 Salamanca, Spain
- Metabolic Engineering Group, Department of Microbiology and Genetics, University of Salamanca, Campus Unamuno, 37007 Salamanca, Spain
| | - Hector Urien
- Instituto de Biología Molecular y Celular del Cancer, Consejo Superior de Investigaciones Científicas – University of Salamanca, Campus Unamuno, 37007 Salamanca, Spain
| | - Arnoud Sonnenberg
- Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Gunnar Jeschke
- Laboratory of Physical Chemistry, ETH Zürich, Vladimir-Prelog-Weg 2, CH-8093 Zürich, Switzerland
| | - José M. de Pereda
- Instituto de Biología Molecular y Celular del Cancer, Consejo Superior de Investigaciones Científicas – University of Salamanca, Campus Unamuno, 37007 Salamanca, Spain
| |
Collapse
|
39
|
Molecular architecture and function of the hemidesmosome. Cell Tissue Res 2014; 360:363-78. [PMID: 25487405 PMCID: PMC4544487 DOI: 10.1007/s00441-014-2061-z] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 11/03/2014] [Indexed: 01/07/2023]
Abstract
Hemidesmosomes are multiprotein complexes that facilitate the stable adhesion of basal epithelial cells to the underlying basement membrane. The mechanical stability of hemidesmosomes relies on multiple interactions of a few protein components that form a membrane-embedded tightly-ordered complex. The core of this complex is provided by integrin α6β4 and P1a, an isoform of the cytoskeletal linker protein plectin that is specifically associated with hemidesmosomes. Integrin α6β4 binds to the extracellular matrix protein laminin-332, whereas P1a forms a bridge to the cytoplasmic keratin intermediate filament network. Other important components are BPAG1e, the epithelial isoform of bullous pemphigoid antigen 1, BPAG2, a collagen-type transmembrane protein and CD151. Inherited or acquired diseases in which essential components of the hemidesmosome are missing or structurally altered result in tissue fragility and blistering. Modulation of hemidesmosome function is of crucial importance for a variety of biological processes, such as terminal differentiation of basal keratinocytes and keratinocyte migration during wound healing and carcinoma invasion. Here, we review the molecular characteristics of the proteins that make up the hemidesmosome core structure and summarize the current knowledge about how their assembly and turnover are regulated by transcriptional and post-translational mechanisms.
Collapse
|
40
|
Wang H, Jin H, Beauvais DM, Rapraeger AC. Cytoplasmic domain interactions of syndecan-1 and syndecan-4 with α6β4 integrin mediate human epidermal growth factor receptor (HER1 and HER2)-dependent motility and survival. J Biol Chem 2014; 289:30318-30332. [PMID: 25202019 PMCID: PMC4215216 DOI: 10.1074/jbc.m114.586438] [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] [Received: 06/03/2014] [Revised: 09/02/2014] [Indexed: 12/14/2022] Open
Abstract
Epithelial cells are highly dependent during wound healing and tumorigenesis on the α6β4 integrin and its association with receptor tyrosine kinases. Previous work showed that phosphorylation of the β4 subunit upon matrix engagement depends on the matrix receptor syndecan (Sdc)-1 engaging the cytoplasmic domain of the β4 integrin and coupling of the integrin to human epidermal growth factor receptor-2 (HER2). In this study, HER2-dependent migration activated by matrix engagement is compared with migration stimulated by EGF. We find that whereas HER2-dependent migration depends on Sdc1, EGF-dependent migration depends on a complex consisting of human epidermal growth factor receptor-1 (HER1, commonly known as EGFR), α6β4, and Sdc4. The two syndecans recognize distinct sites at the extreme C terminus of the β4 integrin cytoplasmic domain. The binding motif in Sdc1 is QEEXYX, composed in part by its syndecan-specific variable (V) region and in part by the second conserved (C2) region that it shares with other syndecans. A cell-penetrating peptide containing this sequence competes for HER2-dependent epithelial migration and carcinoma survival, although it is without effect on the EGFR-stimulated mechanism. β4 mutants bearing mutations specific for Sdc1 and Sdc4 recognition act as dominant negative mutants to block cell spreading or cell migration that depends on HER2 or EGFR, respectively. The interaction of the α6β4 integrin with the syndecans appears critical for it to be utilized as a signaling platform; migration depends on α3β1 integrin binding to laminin 332 (LN332; also known as laminin 5), whereas antibodies that block α6β4 binding are without effect. These findings indicate that specific syndecan family members are likely to have key roles in α6β4 integrin activation by receptor tyrosine kinases.
Collapse
Affiliation(s)
- Haiyao Wang
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Haining Jin
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - DeannaLee M Beauvais
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Alan C Rapraeger
- Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin 53705; Carbone Cancer Center, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, Wisconsin 53705.
| |
Collapse
|
41
|
Chen YH, Chang SH, Wang IJ, Young TH. The mechanism for keratinocyte detaching from pH-responsive chitosan. Biomaterials 2014; 35:9247-54. [PMID: 25129571 DOI: 10.1016/j.biomaterials.2014.07.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 07/19/2014] [Indexed: 01/01/2023]
Abstract
In this study, we compared the detachment ratio of HaCaT and Hs68 cells from pH-responsive chitosan surface by raising medium pH from 7.20 to 7.65 for 60 min. The detachment ratio of elongated Hs68 cells was over 75%, but that of round-shaped HaCaT cells was less than 50%, even extending the incubation time to 6 h or enhancing the cytoskeletal contractile force with the Rho activator CN01. However, the addition of 2 mm of EDTA into the medium at pH 7.65 could effectively detach HaCaT cells (detachment ratio > 90%), indicating that the calcium ion played an important role in the detachment process. Therefore, the family of Ca(+2)-dependent integrin receptors was examined by RT-PCR, real-time PCR and immunocytochemistry. It was found the expression of integrin β4 (ITGb4) was HaCaT cell-specific and the mRNA level of ITGb4 in undetached HaCaT cells was significantly higher than that in detached ones. By modulating ITGb4 activity with specific functional blocking antibody ASC-8, the detachment ratio of HaCaT cells could be increased to be greater than 85%. Conversely, the addition of the ligand of ITGb4 laminin into the culture system decreased the medium pH-induced detachment ratio for HaCaT cells, but not for Hs68 cells. Further addition of ASC-8 could rescue the effect of laminin on preventing the detachment of HaCaT cells from pH-sensitive chitosan surface. Therefore, this study demonstrated the interaction of ITGb4 and laminin played an important role in controlling the detachment of HaCaT cells on pH-responsive chitosan.
Collapse
Affiliation(s)
- Yi-Hsin Chen
- Institute of Polymer Science and Engineering, College of Engineering, National Taiwan University, Taipei, 106, Taiwan
| | - Shao-Hsuan Chang
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, 100, Taiwan
| | - I-Jong Wang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, 100, Taiwan.
| | - Tai-Horng Young
- Institute of Polymer Science and Engineering, College of Engineering, National Taiwan University, Taipei, 106, Taiwan; Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, 100, Taiwan.
| |
Collapse
|
42
|
Ishikawa T, Wondimu Z, Oikawa Y, Gentilcore G, Kiessling R, Egyhazi Brage S, Hansson J, Patarroyo M. Laminins 411 and 421 differentially promote tumor cell migration via α6β1 integrin and MCAM (CD146). Matrix Biol 2014; 38:69-83. [PMID: 24951930 DOI: 10.1016/j.matbio.2014.06.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 06/09/2014] [Accepted: 06/12/2014] [Indexed: 12/23/2022]
Abstract
α4-laminins, such as laminins 411 and 421, are mesenchymal laminins expressed by blood and lymphatic vessels and some tumor cells. Laminin-411 promotes migration of leukocytes and endothelial cells, but the effect of this laminin and laminin-421 on tumor cells is poorly understood. In the present study, we demonstrate that laminin-411 and, to a greater extent, laminin-421 significantly promote migration of tumor cells originated from melanomas, gliomas and different carcinomas via α6β1 integrin. In solid-phase binding assays, both laminins similarly bound α6β1 integrin but only laminin-421, among several laminin isoforms, readily bound MCAM (CD146), a cell-surface adhesion molecule strongly associated with tumor progression. Accordingly, a function-blocking mAb to MCAM inhibited tumor cell migration on laminin-421 but not on laminins 411 or 521. In tumor tissues, melanoma cells co-expressed MCAM, laminin α4, β1, β2 and γ1 chains, and integrin α6 and β1 chains. The present data highlight the novel role of α4-laminins in tumor cell migration and identify laminin-421 as a primary ligand for MCAM and a putative mediator of tumor invasion and metastasis.
Collapse
Affiliation(s)
- Taichi Ishikawa
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Zenebech Wondimu
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yuko Oikawa
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Giusy Gentilcore
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Rolf Kiessling
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - Johan Hansson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Manuel Patarroyo
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
43
|
Laval S, Laklai H, Fanjul M, Pucelle M, Laurell H, Billon-Galés A, Le Guellec S, Delisle MB, Sonnenberg A, Susini C, Pyronnet S, Bousquet C. Dual roles of hemidesmosomal proteins in the pancreatic epithelium: the phosphoinositide 3-kinase decides. Oncogene 2014; 33:1934-44. [PMID: 23624916 DOI: 10.1038/onc.2013.146] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 02/14/2013] [Accepted: 03/11/2013] [Indexed: 02/06/2023]
Abstract
Given the failure of chemo- and biotherapies to fight advanced pancreatic cancer, one major challenge is to identify critical events that initiate invasion. One priming step in epithelia carcinogenesis is the disruption of epithelial cell anchorage to the basement membrane which can be provided by hemidesmosomes (HDs). However, the existence of HDs in pancreatic ductal epithelium and their role in carcinogenesis remain unexplored. HDs have been explored in normal and cancer pancreatic cells, and patient samples. Unique cancer cell models where HD assembly can be pharmacologically manipulated by somatostatin/sst2 signaling have been then used to investigate the role and molecular mechanisms of dynamic HD during pancreatic carcinogenesis. We surprisingly report the presence of mature type-1 HDs comprising the integrin α6β4 and bullous pemphigoid antigen BP180 in the human pancreatic ductal epithelium. Importantly, HDs are shown to disassemble during pancreatic carcinogenesis. HD breakdown requires phosphoinositide 3-kinase (PI3K)-dependent induction of the matrix-metalloprotease MMP-9, which cleaves BP180. Consequently, integrin α6β4 delocalizes to the cell-leading edges where it paradoxically promotes cell migration and invasion through S100A4 activation. As S100A4 in turn stimulates MMP-9 expression, a vicious cycle maintains BP180 cleavage. Inactivation of this PI3K-MMP-9-S100A4 signaling loop conversely blocks BP180 cleavage, induces HD reassembly and inhibits cell invasion. We conclude that mature type-1 HDs are critical anchoring structures for the pancreatic ductal epithelium whose disruption, upon PI3K activation during carcinogenesis, provokes pancreatic cancer cell migration and invasion.
Collapse
Affiliation(s)
- S Laval
- 1] INSERM UMR 1037, Laboratoire d'excellence Toulouse Cancer (labex TOUCAN), Equipe labellisée Ligue Nationale Contre le Cancer (LNCC), Centre de Recherche en Cancérologie de Toulouse (CRCT), Toulouse, France [2] Université Toulouse III Paul Sabatier, Toulouse, France
| | - H Laklai
- 1] INSERM UMR 1037, Laboratoire d'excellence Toulouse Cancer (labex TOUCAN), Equipe labellisée Ligue Nationale Contre le Cancer (LNCC), Centre de Recherche en Cancérologie de Toulouse (CRCT), Toulouse, France [2] Université Toulouse III Paul Sabatier, Toulouse, France
| | - M Fanjul
- 1] INSERM UMR 1037, Laboratoire d'excellence Toulouse Cancer (labex TOUCAN), Equipe labellisée Ligue Nationale Contre le Cancer (LNCC), Centre de Recherche en Cancérologie de Toulouse (CRCT), Toulouse, France [2] Université Toulouse III Paul Sabatier, Toulouse, France
| | - M Pucelle
- INSERM UMR 1037, Laboratoire d'excellence Toulouse Cancer (labex TOUCAN), Equipe labellisée Ligue Nationale Contre le Cancer (LNCC), Centre de Recherche en Cancérologie de Toulouse (CRCT), Toulouse, France
| | - H Laurell
- 1] INSERM UMR 1037, Laboratoire d'excellence Toulouse Cancer (labex TOUCAN), Equipe labellisée Ligue Nationale Contre le Cancer (LNCC), Centre de Recherche en Cancérologie de Toulouse (CRCT), Toulouse, France [2] Université Toulouse III Paul Sabatier, Toulouse, France
| | - A Billon-Galés
- 1] INSERM UMR 1037, Laboratoire d'excellence Toulouse Cancer (labex TOUCAN), Equipe labellisée Ligue Nationale Contre le Cancer (LNCC), Centre de Recherche en Cancérologie de Toulouse (CRCT), Toulouse, France [2] Université Toulouse III Paul Sabatier, Toulouse, France
| | - S Le Guellec
- Services d'Anatomie et Cytologie Pathologique of Hôpital Rangueil, Toulouse, France
| | - M-B Delisle
- Services d'Anatomie et Cytologie Pathologique of Hôpital Rangueil, Toulouse, France
| | - A Sonnenberg
- Department of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - C Susini
- 1] INSERM UMR 1037, Laboratoire d'excellence Toulouse Cancer (labex TOUCAN), Equipe labellisée Ligue Nationale Contre le Cancer (LNCC), Centre de Recherche en Cancérologie de Toulouse (CRCT), Toulouse, France [2] Université Toulouse III Paul Sabatier, Toulouse, France
| | - S Pyronnet
- 1] INSERM UMR 1037, Laboratoire d'excellence Toulouse Cancer (labex TOUCAN), Equipe labellisée Ligue Nationale Contre le Cancer (LNCC), Centre de Recherche en Cancérologie de Toulouse (CRCT), Toulouse, France [2] Université Toulouse III Paul Sabatier, Toulouse, France
| | - C Bousquet
- 1] INSERM UMR 1037, Laboratoire d'excellence Toulouse Cancer (labex TOUCAN), Equipe labellisée Ligue Nationale Contre le Cancer (LNCC), Centre de Recherche en Cancérologie de Toulouse (CRCT), Toulouse, France [2] Université Toulouse III Paul Sabatier, Toulouse, France
| |
Collapse
|
44
|
Vijayakumar S, Dang S, Marinkovich MP, Lazarova Z, Yoder B, Torres VE, Wallace DP. Aberrant expression of laminin-332 promotes cell proliferation and cyst growth in ARPKD. Am J Physiol Renal Physiol 2014; 306:F640-54. [PMID: 24370592 PMCID: PMC3949036 DOI: 10.1152/ajprenal.00104.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 12/23/2013] [Indexed: 11/22/2022] Open
Abstract
Basement membrane abnormalities have often been observed in kidney cysts of polycystic kidney disease (PKD) patients and animal models. There is an abnormal deposition of extracellular matrix molecules, including laminin-α3,β3,γ2 (laminin-332), in human autosomal dominant PKD (ADPKD). Knockdown of PKD1 paralogs in zebrafish leads to dysregulated synthesis of the extracellular matrix, suggesting that altered basement membrane assembly may be a primary defect in ADPKD. In this study, we demonstrate that laminin-332 is aberrantly expressed in cysts and precystic tubules of human autosomal recessive PKD (ARPKD) kidneys as well as in the kidneys of PCK rats, an orthologous ARPKD model. There was aberrant expression of laminin-γ2 as early as postnatal day 2 and elevated laminin-332 protein in postnatal day 30, coinciding with the formation and early growth of renal cysts in PCK rat kidneys. We also show that a kidney cell line derived from Oak Ridge polycystic kidney mice, another model of ARPKD, exhibited abnormal lumen-deficient and multilumen structures in Matrigel culture. These cells had increased proliferation rates and altered expression levels of laminin-332 compared with their rescued counterparts. A function-blocking polyclonal antibody to laminin-332 significantly inhibited their abnormal proliferation rates and rescued their aberrant phenotype in Matrigel culture. Furthermore, abnormal laminin-332 expression in cysts originating from collecting ducts and proximal tubules as well as in precystic tubules was observed in a human end-stage ADPKD kidney. Our results suggest that abnormal expression of laminin-332 contributes to the aberrant proliferation of cyst epithelial cells and cyst growth in genetic forms of PKD.
Collapse
Affiliation(s)
- Soundarapandian Vijayakumar
- Dept. of Natural Sciences and Mathematics, SUNY Cobleskill, 111 Schenectady Ave. WH200, Cobleskill, NY 12043.
| | | | | | | | | | | | | |
Collapse
|
45
|
Chen Q, Xu R, Zeng C, Lu Q, Huang D, Shi C, Zhang W, Deng L, Yan R, Rao H, Gao G, Luo S. Down-regulation of Gli transcription factor leads to the inhibition of migration and invasion of ovarian cancer cells via integrin β4-mediated FAK signaling. PLoS One 2014; 9:e88386. [PMID: 24533083 PMCID: PMC3922814 DOI: 10.1371/journal.pone.0088386] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 01/06/2014] [Indexed: 01/25/2023] Open
Abstract
Background Recent evidence suggests that aberrant activation of Hedgehog (Hh) signaling by Gli transcription factors is characteristic of a variety of aggressive human carcinomas including ovarian cancer. Therefore, chemotherapeutic agents that inhibit activation of Gli transcription factors have emerged as promising novel therapeutic drugs for ovarian cancer. Results In this study, we show that activation of Hh signaling promoted cellular migration and invasion, whereas blockade of Hh signaling with GANT61 suppressed cellular migration and invasion in ovarian cancer cells. After treatment with GANT61, cDNA microarray analyses revealed changes in many genes such as Integrin β4 subunit (ITGB4), focal adhesion kinase (FAK), etc. Furthermore, ITGB4 expression was up-regulated by Sonic Hedgehog (Shh) ligand and down-regulated by Hh signaling inhibitor. The Shh-mediated ovarian cell migration and invasion was blocked by neutralizing antibodies to ITGB4. In addition, phosphorylations of FAK were increased by Shh and decreased by Hh signaling inhibitor. Inhibition of Gli1 expression using siRNA mimicked the effects of GANT61 treatment, supporting the specificity of GANT61. Further investigations showed that activation of FAK was required for Shh-mediated cell migration and invasion. Finally, we found that down-regulation of Gli reduced the expression of ITGB4 and the phosphorylated FAK, resulting in the inhibition of tumor growth in vivo. Conclusions The Hh signaling pathway induces cell migration and invasion through ITGB4-mediated activation of FAK in ovarian cancer. Our findings suggest that the diminishment of crosstalk between phosphorylated FAK and ITGB4 due to the down-regulation of Gli family transcription factors might play a pivotal role for inhibiting ovarian cancer progression.
Collapse
Affiliation(s)
- Qi Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Rong Xu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Chunyan Zeng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Quqin Lu
- Department of Biostatistics & Epidemiology, School of Public Health, Nanchang University, Nanchang, Jiangxi, China
| | - Dengliang Huang
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Chao Shi
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Weilong Zhang
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Libin Deng
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Runwei Yan
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Hai Rao
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Guolan Gao
- Department of Obstetrics and Gynecology, General Hospital of Beijing Aeronautics, Beijing, China
- * E-mail: (SL); (GG)
| | - Shiwen Luo
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- * E-mail: (SL); (GG)
| |
Collapse
|
46
|
Beauséjour M, Thibodeau S, Demers MJ, Bouchard V, Gauthier R, Beaulieu JF, Vachon PH. Suppression of anoikis in human intestinal epithelial cells: differentiation state-selective roles of α2β1, α3β1, α5β1, and α6β4 integrins. BMC Cell Biol 2013; 14:53. [PMID: 24289209 PMCID: PMC4219346 DOI: 10.1186/1471-2121-14-53] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 11/26/2013] [Indexed: 12/26/2022] Open
Abstract
Background Regulation of anoikis in human intestinal epithelial cells (IECs) implicates differentiation state-specific mechanisms. Human IECs express distinct repertoires of integrins according to their state of differentiation. Therefore, we investigated whether α2β1, α3β1, α5β1, and α6β4 integrins perform differentiation state-specific roles in the suppression of IEC anoikis. Results Human (HIEC, Caco-2/15) IECs were exposed to specific antibodies that block the binding activity of integrin subunits (α2, α3, α5, α6, β1 or β4) to verify whether or not their inhibition induced anoikis. The knockdown of α6 was also performed by shRNA. Additionally, apoptosis/anoikis was induced by pharmacological inhibition of Fak (PF573228) or Src (PP2). Anoikis/apoptosis was assayed by DNA laddering, ISEL, and/or caspase activity (CASP-8, -9, or -3). Activation levels of Fak and Src, as well as functional Fak-Src interactions, were also assessed. We report herein that differentiated IECs exhibit a greater sensitivity to anoikis than undifferentiated ones. This involves an earlier onset of anoikis when kept in suspension, as well as significantly greater contributions from β1 and β4 integrins in the suppression of anoikis in differentiated cells, and functional distinctions between β1 and β4 integrins in engaging both Fak and Src, or Src only, respectively. Likewise, Fak performs significantly greater contributions in the suppression of anoikis in differentiated cells. Additionally, we show that α2β1 and α5β1 suppress anoikis in undifferentiated cells, whereas α3β1 does so in differentiated ones. Furthermore, we provide evidence that α6β4 contributes to the suppression of anoikis in a primarily α6 subunit-dependent manner in undifferentiated cells, whereas this same integrin in differentiated cells performs significantly greater contributions in anoikis suppression than its undifferentiated state-counterpart, in addition to doing so through a dependence on both of its subunits. Conclusions Our findings indicate that the suppression of human IEC anoikis implicates differentiation state-selective repertoires of integrins, which in turn results into distinctions in anoikis regulation, and sensitivity, between undifferentiated and differentiated IECs. These data further the functional understanding of the concept that the suppression of anoikis is subjected to cell differentiation state-selective mechanisms.
Collapse
Affiliation(s)
- Marco Beauséjour
- Département d'anatomie et de biologie cellulaire, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, J1H5N4 Sherbrooke, Québec, Canada.
| | | | | | | | | | | | | |
Collapse
|
47
|
Thuma F, Ngora H, Zöller M. The metastasis-associated molecule C4.4A promotes tissue invasion and anchorage independence by associating with the alpha6beta4 integrin. Mol Oncol 2013; 7:917-28. [PMID: 23727360 PMCID: PMC5528461 DOI: 10.1016/j.molonc.2013.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 05/01/2013] [Accepted: 05/02/2013] [Indexed: 01/13/2023] Open
Abstract
C4.4A is a metastasis-associated molecule that functions appear to rely on associated alph6beta4 integrin. To corroborate the impact of the C4.4A-alpha6beta4 integrin association on metastasis formation, C4.4A was knocked-down in a highly metastatic rat pancreatic adenocarcinoma (ASML, ASML-C4.4Akd). Metastasis formation by ASML-C4.4Akd cells after intrafootpad application was strongly retarded in draining nodes and lung colonization was rare. Furthermore, cisplatin treatment significantly prolonged the survival time only of ASML-C4.4Akd-bearing rats. ASML-C4.4Akd cells display reduced migratory activity and impaired matrix protein degradation due to inefficient MMP14 activation; loss of drug-resistance is due to mitigated PI3K/Akt pathway activation. These losses of function rely on the laminin receptor C4.4A recruiting activated alpha6beta4 integrin into rafts, where C4.4A cooperates with alpha6beta4 and via alpha6beta4 with MMP14. Within this raft-located complex, MMP14 provokes focalized matrix degradation and mostly alpha6beta4 integrin promotes BAD phosphorylation and upregulated Bcl2 and BclXl expression. Thus, metastasis-promoting activities of C4.4A are not genuine characteristics of C4.4A. Instead, the raft-located laminin receptor C4.4A recruits alpha6beta4 integrin and supports via the alpha6beta4 integrin MMP14 activation. Thereby C4.4A acts as a linker to facilitate several steps in the metastatic cascade. Taking the restricted C4.4A expression in non-transformed tissue, this knowledge should pave the way toward the use of C4.4A as a therapeutic target.
Collapse
Affiliation(s)
- Florian Thuma
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg D-69120, Germany
| | - Honoré Ngora
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg D-69120, Germany
| | - Margot Zöller
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg D-69120, Germany
- German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
48
|
Yamada T, Kuramitsu K, Rikitsu E, Kurita S, Ikeda W, Takai Y. Nectin and junctional adhesion molecule are critical cell adhesion molecules for the apico-basal alignment of adherens and tight junctions in epithelial cells. Genes Cells 2013; 18:985-98. [PMID: 24112238 DOI: 10.1111/gtc.12091] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 07/30/2013] [Indexed: 02/02/2023]
Abstract
Tight junctions (TJs) and adherens junctions (AJs) form an apical junctional complex at the apical side of the lateral membranes of epithelial cells, in which TJs are aligned at the apical side of AJs. Many cell adhesion molecules (CAMs) and cell polarity molecules (CPMs) cooperatively regulate the formation of the apical junctional complex, but the mechanism for the alignment of TJs at the apical side of AJs is not fully understood. We developed a cellular system with which epithelial-like TJs and AJs were reconstituted in fibroblasts and analyzed the cooperative roles of CAMs and CPMs. We exogenously expressed various combinations of CAMs and CPMs in fibroblasts that express negligible amounts of these molecules endogenously. In these cells, the nectin-based cell-cell adhesion was formed at the apical side of the junctional adhesion molecule (JAM)-based cell-cell adhesion, and cadherin and claudin were recruited to the nectin-3- and JAM-based cell-cell adhesion sites to form AJ-like and TJ-like domains, respectively. This inversed alignment of the AJ-like and TJ-like domains was reversed by complementary expression of CPMs Par-3, atypical protein kinase C, Par-6, Crb3, Pals1 and Patj. We describe the cooperative roles of these CAMs and CPMs in the apico-basal alignment of TJs and AJs in epithelial cells.
Collapse
Affiliation(s)
- Tomohiro Yamada
- KAN Research Institute, Inc., 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | | | | | | | | | | |
Collapse
|
49
|
Zhou B, Gibson-Corley KN, Herndon ME, Sun Y, Gustafson-Wagner E, Teoh-Fitzgerald M, Domann FE, Henry MD, Stipp CS. Integrin α3β1 can function to promote spontaneous metastasis and lung colonization of invasive breast carcinoma. Mol Cancer Res 2013; 12:143-154. [PMID: 24002891 DOI: 10.1158/1541-7786.mcr-13-0184] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
UNLABELLED Significant evidence implicates α3β1 integrin in promoting breast cancer tumorigenesis and metastasis-associated cell behaviors in vitro and in vivo. However, the extent to which α3β1 is actually required for breast cancer metastasis remains to be determined. We used RNA interference to silence α3 integrin expression by approximately 70% in 4T1 murine mammary carcinoma cells, a model of aggressive, metastatic breast cancer. Loss of α3 integrin reduced adhesion, spreading, and proliferation on laminin isoforms, and modestly reduced the growth of orthotopically implanted cells. However, spontaneous metastasis to lung was strikingly curtailed. Experimental lung colonization after tail vein injection revealed a similar loss of metastatic capacity for the α3-silenced (α3si) cells, suggesting that critical, α3-dependent events at the metastatic site could account for much of α3β1's contribution to metastasis in this model. Reexpressing α3 in the α3si cells reversed the loss of metastatic capacity, and silencing another target, the small GTPase RhoC, had no effect, supporting the specificity of the effect of silencing α3. Parental, α3si, and α3-rescued cells, all secreted abundant laminin α5 (LAMA5), an α3β1 integrin ligand, suggesting that loss of α3 integrin might disrupt an autocrine loop that could function to sustain metastatic growth. Analysis of human breast cancer cases revealed reduced survival in cases where α3 integrin and LAMA5 are both overexpressed. IMPLICATIONS α3 integrin or downstream effectors may be potential therapeutic targets in disseminated breast cancers, especially when laminin α5 or other α3 integrin ligands are also over-expressed.
Collapse
Affiliation(s)
- Bo Zhou
- Department of Biology, University of Iowa, Iowa City, IA, 52242 USA
| | | | - Mary E Herndon
- Department of Biology, University of Iowa, Iowa City, IA, 52242 USA
| | - Yihan Sun
- Department of Biology, University of Iowa, Iowa City, IA, 52242 USA
| | | | - Melissa Teoh-Fitzgerald
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA, 52242 USA
| | - Frederick E Domann
- Department of Pathology, University of Iowa, Iowa City, IA, 52242 USA.,Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA, 52242 USA.,Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, 52242 USA
| | - Michael D Henry
- Department of Pathology, University of Iowa, Iowa City, IA, 52242 USA.,Department of Molecular Physiology & Biophysics, University of Iowa, Iowa City, IA, 52242 USA.,Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, 52242 USA
| | - Christopher S Stipp
- Department of Biology, University of Iowa, Iowa City, IA, 52242 USA.,Department of Molecular Physiology & Biophysics, University of Iowa, Iowa City, IA, 52242 USA.,Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, 52242 USA
| |
Collapse
|
50
|
Carubbi C, Gobbi G, Bucci G, Gesi M, Vitale M, Mirandola P. Skin, Inflammation and Sulfurous Waters: What is Known, What is Believed. EUR J INFLAMM 2013. [DOI: 10.1177/1721727x1301100303] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
One could argue that balneotherapy and mud therapy would have not lasted 2,000 years or so If they were not effective. No doubt a long history cannot be taken per se as scientific proof of efficacy. Some empiricism is still present in the field: the concept of spa itself is quite confounding, whereas spring waters are used for leisure purposes but also for non-acute patient therapy and late phases of clinical recovery. These confounding elements ultimately feed the opinion of those who aprioristically reject any potential beneficial effect of balneotherapy: instead, it should at least generate questions that deserve scientific answers. Clinical practices sequentially integrating pharmacological therapy with those natural principles for which a sufficient scientific demonstration is available, would probably cut the costs of public health, generating widespread advantages for the community. Recently, it has become evident that mineral waters may have intrinsic pharmacological properties. Of the numerous salts dissolved in thermal waters that might show pharmacological properties, for certain hydrogen sulfide (H2S) contained in sulfurous waters is the one that has obtained greater scientific attention, to which should be added the extensive scientific effort recently dedicated to H2S as a cellular gasotransmitter, independently from its natural sources. Dermatology and cosmetics are among the most studied applications of sulfurous waters, around which, however, some empiricism still confounds opinions: we therefore considered that a state-of-the-art focus on this topic might be timely and useful for future studies.
Collapse
Affiliation(s)
- C. Carubbi
- Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Parma, Italy
| | - G. Gobbi
- Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Parma, Italy
| | - G. Bucci
- Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Parma, Italy
| | - M. Gesi
- Department of Human Morphology and Applied Biology, University of Pisa, Pisa, Italy
| | - M. Vitale
- Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Parma, Italy
- Scientific Coordinator of the Italian Foundation for Scientific Research in Balneotherapy (FoRST), Rome, Italy
| | - P. Mirandola
- Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T.), University of Parma, Parma, Italy
| |
Collapse
|