1
|
Wu Y, You Y, Jiang T, He Y, Fan Q, Zeng X, Li T, Lu Y, Qi L, Zhou F, Sun L, Wang D, Zou Y, Zhang G, Yuan Y, Mao Y. Proximity Labeling and Genetic Screening Reveal that DSG2 is a Counter Receptor of Siglec-9 and Suppresses Macrophage Phagocytosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2406654. [PMID: 39813162 DOI: 10.1002/advs.202406654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 12/23/2024] [Indexed: 01/18/2025]
Abstract
Cancer cells present sialylated glycoconjugates that modulate the activity of various immune cells within the tumor microenvironment through trans interaction with immunosuppressive Siglec receptors. Identifying counter receptors for Siglecs can provide valuable targets for cancer immunotherapy, but it presents significant challenges. Here, the identification of DSG2 (Desmoglein 2) as a dominant counter receptor of Siglec-9 in melanoma cells is reported, using a workflow that combines the strength of proximity labeling and the advantage of CRISPR knockout screening. It is further demonstrated that the interaction between DSG2 and Siglec-9 is mainly dependent on sialic acid-bearing N-glycans on DSG2. Importantly, blocking trans interaction between DSG2 and Siglec-9 significantly enhances macrophage phagocytosis of melanoma cells and, to a lesser extent, other cancer cells. The work thus suggests sialylated DSG2 as a potential "don't eat me" signal molecule with therapeutic potentials in cancer immunotherapy.
Collapse
Affiliation(s)
- Ying Wu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yuyu You
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Tingsong Jiang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yuqi He
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Qingchi Fan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Xinlei Zeng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ting Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yuxing Lu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Liang Qi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Fengxia Zhou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Lingyu Sun
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Danyang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510275, China
| | - Yong Zou
- Department of Blood Transfusion, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, China
| | - Guigen Zhang
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yanqiu Yuan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yang Mao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, Guangzhou, 510006, China
| |
Collapse
|
2
|
Luo Q, Li X, Xie K. Plakophilin 1 in carcinogenesis. Mol Carcinog 2024; 63:1855-1865. [PMID: 38888207 DOI: 10.1002/mc.23779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/11/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Abstract
Plakophilin 1 (PKP1) belongs to the desmosome family as an anchoring junction protein in cellular junctions. It localizes at the interface of the cell membrane and cytoplasm. Although PKP1 is a non-transmembrane protein, it may become associated with the cell membrane via transmembrane proteins such as desmocollins and desmogleins. Homozygous deletion of PKP1 results in ectodermal dysplasia-skin fragility syndrome (EDSF) and complete knockout of PKP1 in mice produces comparable symptoms to EDSF in humans, although mice do not survive more than 24 h. PKP1 is not limited to expression in desmosomal structures, but is rather widely expressed in cytoplasm and nucleus, where it assumes important cellular functions. This review will summarize distinct roles of PKP1 in the cell membrane, cytoplasm, and nucleus with an overview of relevant studies on its function in diverse types of cancer.
Collapse
Affiliation(s)
- Qiang Luo
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, Guangdong, China
| | - Xiaojia Li
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, Guangdong, China
| | - Keping Xie
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, Guangdong, China
- The Second Affiliated Hospital and Guangzhou First People's Hospital, South China University of Technology School of Medicine, Guangzhou, Guangdong, China
- The South China University of Technology Comprehensive Cancer Center, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Diede C, Walker T, Carr DR, Shahwan KT. Grading differentiation in cutaneous squamous cell carcinoma: a review of the literature. Arch Dermatol Res 2024; 316:434. [PMID: 38935165 DOI: 10.1007/s00403-024-03184-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 05/23/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024]
Abstract
Poor differentiation is strongly associated with poor outcomes in cutaneous squamous cell carcinoma (CSCC). In addition, the National Comprehensive Cancer Network (NCCN) guidelines designate poorly differentiated tumors as "very high risk". Despite its clear prognostic implications, there is no standardized grading system for CSCC differentiation in common use today. CSCC differentiation is graded inconsistently by both dermatopathologists and Mohs surgeons, and reliability studies have demonstrated suboptimal inter- and intra-rater reliability in both of these groups. The absence of a standardized and reliable grading system has impeded the use of differentiation in CSCC staging, despite its apparent correlation with disease outcomes. We performed a comprehensive review of the literature summarizing historical CSCC differentiation grading systems, as well as grading systems in non-cutaneous head and neck SCC as a point of reference. Relevant articles were identified by searching Embase and PubMed, as well as by reviewing reference lists for additional articles and histology textbook excerpts. CSCC grading systems that were identified and summarized include the historical Broders system, the World Health Organization system, the College of American Pathologists' system, and a system described by a 2023 Delphi consensus panel of dermatopathologists.
Collapse
Affiliation(s)
- Claire Diede
- University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Trent Walker
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - David R Carr
- Department of Dermatology, The Ohio State University Medical Center, 6700 University Blvd, Columbus, OH, 43016, USA
| | - Kathryn T Shahwan
- University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, USA.
- Department of Dermatology, The Ohio State University Medical Center, 6700 University Blvd, Columbus, OH, 43016, USA.
- Department of Dermatology, Altru Health System, Grand Forks, Grand Forks, ND, USA.
| |
Collapse
|
4
|
Myo Min KK, Ffrench CB, McClure BJ, Ortiz M, Dorward EL, Samuel MS, Ebert LM, Mahoney MG, Bonder CS. Desmoglein-2 as a cancer modulator: friend or foe? Front Oncol 2023; 13:1327478. [PMID: 38188287 PMCID: PMC10766750 DOI: 10.3389/fonc.2023.1327478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Desmoglein-2 (DSG2) is a calcium-binding single pass transmembrane glycoprotein and a member of the large cadherin family. Until recently, DSG2 was thought to only function as a cell adhesion protein embedded within desmosome junctions designed to enable cells to better tolerate mechanical stress. However, additional roles for DSG2 outside of desmosomes are continuing to emerge, particularly in cancer. Herein, we review the current literature on DSG2 in cancer and detail its impact on biological functions such as cell adhesion, proliferation, migration, invasion, intracellular signaling, extracellular vesicle release and vasculogenic mimicry. An increased understanding of the diverse repertoire of the biological functions of DSG2 holds promise to exploit this cell surface protein as a potential prognostic biomarker and/or target for better patient outcomes. This review explores the canonical and non-canonical functions of DSG2, as well as the context-dependent impacts of DSG2 in the realm of cancer.
Collapse
Affiliation(s)
- Kay K. Myo Min
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Charlie B. Ffrench
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Barbara J. McClure
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Michael Ortiz
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Emma L. Dorward
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Michael S. Samuel
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Basil Hetzel Institute, Queen Elizabeth Hospital, SA, Adelaide, Australia
| | - Lisa M. Ebert
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
- Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Mỹ G. Mahoney
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Claudine S. Bonder
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
5
|
Chen S, Hu S, Zhou B, Cheng B, Tong H, Su D, Li X, Chen Y, Zhang G. Telomere-related prognostic biomarkers for survival assessments in pancreatic cancer. Sci Rep 2023; 13:10586. [PMID: 37391503 PMCID: PMC10313686 DOI: 10.1038/s41598-023-37836-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/28/2023] [Indexed: 07/02/2023] Open
Abstract
Human telomeres are linked to genetic instability and a higher risk of developing cancer. Therefore, to improve the dismal prognosis of pancreatic cancer patients, a thorough investigation of the association between telomere-related genes and pancreatic cancer is required. Combat from the R package "SVA" was performed to correct the batch effects between the TCGA-PAAD and GTEx datasets. After differentially expressed genes (DEGs) were assessed, we constructed a prognostic risk model through univariate Cox regression, LASSO-Cox regression, and multivariate Cox regression analysis. Data from the ICGC, GSE62452, GSE71729, and GSE78229 cohorts were used as test cohorts for validating the prognostic signature. The major impact of the signature on the tumor microenvironment and its response to immune checkpoint drugs was also evaluated. Finally, PAAD tissue microarrays were fabricated and immunohistochemistry was performed to explore the expression of this signature in clinical samples. After calculating 502 telomere-associated DEGs, we constructed a three-gene prognostic signature (DSG2, LDHA, and RACGAP1) that can be effectively applied to the prognostic classification of pancreatic cancer patients in multiple datasets, including TCGA, ICGC, GSE62452, GSE71729, and GSE78229 cohorts. In addition, we have screened a variety of tumor-sensitive drugs targeting this signature. Finally, we also found that protein levels of DSG2, LDHA, and RACGAP1 were upregulated in pancreatic cancer tissues compared to normal tissues by immunohistochemistry analysis. We established and validated a telomere gene-related prognostic signature for pancreatic cancer and confirmed the upregulation of DSG2, LDHA, and RACGAP1 expression in clinical samples, which may provide new ideas for individualized immunotherapy.
Collapse
Affiliation(s)
- Shengyang Chen
- Department of Hepatobiliary Pancreatic Surgery, Zhengzhou University Fifth Affiliated Hospital, Kangfu Front Street 3#, Zhengzhou, 450052, China.
| | - Shuiquan Hu
- Department of Hepatobiliary Pancreatic Surgery, Zhengzhou University Fifth Affiliated Hospital, Kangfu Front Street 3#, Zhengzhou, 450052, China
| | - Baizhong Zhou
- Department of Hepatobiliary Pancreatic Surgery, Zhengzhou University Fifth Affiliated Hospital, Kangfu Front Street 3#, Zhengzhou, 450052, China
| | - Bingbing Cheng
- Department of Hepatobiliary Pancreatic Surgery, Zhengzhou University Fifth Affiliated Hospital, Kangfu Front Street 3#, Zhengzhou, 450052, China
| | - Hao Tong
- Department of Hepatobiliary Pancreatic Surgery, Zhengzhou University Fifth Affiliated Hospital, Kangfu Front Street 3#, Zhengzhou, 450052, China
| | - Dongchao Su
- Department of Hepatobiliary Pancreatic Surgery, Zhengzhou University Fifth Affiliated Hospital, Kangfu Front Street 3#, Zhengzhou, 450052, China
| | - Xiaoyong Li
- Department of Hepatobiliary Pancreatic Surgery, Zhengzhou University Fifth Affiliated Hospital, Kangfu Front Street 3#, Zhengzhou, 450052, China
| | - Yanjun Chen
- Department of Hepatobiliary Pancreatic Surgery, Zhengzhou University Fifth Affiliated Hospital, Kangfu Front Street 3#, Zhengzhou, 450052, China
| | - Genhao Zhang
- Department of Blood Transfusion, Zhengzhou University First Affiliated Hospital, Zhengzhou, China
| |
Collapse
|
6
|
Pogorzelska-Dyrbus J, Szepietowski JC. Adhesion Molecules in Non-melanoma Skin Cancers: A Comprehensive Review. In Vivo 2021; 35:1327-1336. [PMID: 33910810 DOI: 10.21873/invivo.12385] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/05/2021] [Accepted: 03/19/2021] [Indexed: 12/18/2022]
Abstract
Basal cell carcinoma (BCC) and squamous cell carcinoma (SCC) are the most frequently diagnosed cancers, generating significant medical and financial problems. Cutaneous carcinogenesis is a very complex process characterized by genetic and molecular alterations, and mediated by various proteins and pathways. Cell adhesion molecules (CAMs) are transmembrane proteins responsible for cell-to-cell and cell-to-extracellular matrix adhesion, engaged in all steps of tumor progression. Based on their structures they are divided into five major groups: cadherins, integrins, selectins, immunoglobulins and CD44 family. Cadherins, integrins and CD44 are the most studied in the context of non-melanoma skin cancers. The differences in expression of adhesion molecules may be related to the invasiveness of these tumors, through the loss of tissue integrity, neovascularization and alterations in intercellular signaling processes. In this article, each group of CAMs is briefly described and the present knowledge on their role in the development of non-melanoma skin cancers is summarized.
Collapse
Affiliation(s)
| | - Jacek C Szepietowski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
7
|
Weng CF, Huang CJ, Wu MH, Lee HHC, Ling TY. Co-Expression of Coxsackievirus/Adenovirus Receptors and Desmoglein 2 in Lung Adenocarcinoma: A Comprehensive Analysis of Bioinformatics and Tissue Microarrays. J Clin Med 2020; 9:jcm9113693. [PMID: 33217893 PMCID: PMC7698609 DOI: 10.3390/jcm9113693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/06/2020] [Accepted: 11/16/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction: Coxsackievirus/adenovirus receptors (CARs) and desmoglein-2 (DSG2) are similar molecules to adenovirus-based vectors in the cell membrane. They have been found to be associated with lung epithelial cell tumorigenesis and can be useful markers in predicting survival outcome in lung adenocarcinoma (LUAD). Methods: A gene ontology enrichment analysis disclosed that DSG2 was highly correlated with CAR. Survival analysis was then performed on 262 samples from the Cancer Genome Atlas, forming “Stage 1A” or “Stage 1B”. We therefore analyzed a tissue microarray (TMA) comprised of 108 lung samples and an immunohistochemical assay. Computer counting software was used to calculate the H-score of the immune intensity. Cox regression and Kaplan–Meier analyses were used to determine the prognostic value. Results: CAR and DSG2 genes are highly co-expressed in early stage LUAD and associated with significantly poorer survival (p = 0.0046). TMA also showed that CAR/DSG2 expressions were altered in lung cancer tissue. CAR in the TMA was correlated with proliferation, apoptosis, and epithelial–mesenchymal transition (EMT), while DSG2 was associated with proliferation only. The Kaplan–Meier survival analysis revealed that CAR, DSG2, or a co-expression of CAR/DSG2 was associated with poorer overall survival. Conclusions: The co-expression of CAR/DSG2 predicted a worse overall survival in LUAD. CAR combined with DSG2 expression can predict prognosis.
Collapse
Affiliation(s)
- Ching-Fu Weng
- Division of Pulmonary Medicine, Department of Internal Medicine, Hsinchu Cathay General Hospital, Hsinchu 300, Taiwan;
- Department and Graduate Institute of Pharmacology, National Taiwan University, Taipei 100, Taiwan
| | - Chi-Jung Huang
- Medical Research Center, Cathay General Hospital, Taipei 106, Taiwan;
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan
| | - Mei-Hsuan Wu
- Teaching and Research Center, Hsinchu Cathay General Hospital, Hsinchu 300, Taiwan;
| | - Henry Hsin-Chung Lee
- School of Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan
- Department of Surgery, Hsinchu Cathay General Hospital, Hsinchu 300, Taiwan
- Graduate Institute of Translational and Interdisciplinary Medicine, College of Health Sciences and Technology, National Central University, Taoyuan 320, Taiwan
- Correspondence: (H.H.-C.L.); (T.-Y.L.); Tel.: +886-3-527-8999 (ext. 61346) (H.H.-C.L.); +886-2-2312-3456 (ext. 88322) (T.-Y.L.)
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, National Taiwan University, Taipei 100, Taiwan
- Correspondence: (H.H.-C.L.); (T.-Y.L.); Tel.: +886-3-527-8999 (ext. 61346) (H.H.-C.L.); +886-2-2312-3456 (ext. 88322) (T.-Y.L.)
| |
Collapse
|
8
|
Qin S, Liao Y, Du Q, Wang W, Huang J, Liu P, Shang C, Liu T, Xia M, Yao S. DSG2 expression is correlated with poor prognosis and promotes early-stage cervical cancer. Cancer Cell Int 2020; 20:206. [PMID: 32514251 PMCID: PMC7268232 DOI: 10.1186/s12935-020-01292-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/25/2020] [Indexed: 12/15/2022] Open
Abstract
Background The pathogenesis and developmental mechanism of early-stage (FIGO 2009 IA2-IIA2) cervical cancer (CC) remain unclear. Seeking novel molecular biomarkers based on The Cancer Genome Atlas (TCGA) will facilitate the understanding of CC pathogenesis and help evaluate early-stage CC prognosis. Methods To identify prognosis-related genes in early-stage CC, we analyzed TCGA mRNA-seq data and clinical data by univariate Cox and Kaplan-Meier plotter analyses. Differential expression analysis identified upregulated genes in early-stage CC. Combined with the genes correlated with unfavorable prognosis, we selected desmoglein-2 (DSG2) for further investigation. To detect DSG2 expression in early-stage CC, we used immunohistochemistry (IHC), quantitative real-time PCR (qRT-PCR) and western blotting. The relationship between the expression of DSG2 and clinical features was analyzed by the Chi square test. Cox analysis was applied to assess the relationship between CC overall survival (OS) and risk factors. The correlations between DSG2 expression and CC cell line proliferation and migration were investigated with Cell Counting Kit-8 (CCK-8) and migration assays. Results There were 416 prognosis-related genes in early-stage CC. DSG2, matrix metallopeptidase 1 (MMP1), carbonic anhydrase IX (CA9), homeobox A1 (HOXA1), and serine protease inhibitor B3 (SERPINB3) were upregulated in early-stage CC compared with adjacent noncancerous tissue (ANT) and correlated with unfavorable prognosis. Among them, DSG2 was most significantly correlated with patient survival. Coexpression analysis indicated that DSG2 was probably involved in cell division, positive regulation of transferase activity, positive regulation of cell migration, EGFR upregulation pathway and regulation of lymphangiogenesis. IHC, qRT-PCR and western blotting showed that DSG2 expression was higher in CC than in normal tissue. Significant correlations were identified between DSG2 expression and several aggressive clinical features, including pelvic lymph node metastasis (PLNM). Multivariate Cox analysis showed that DSG2 and PLNM were independent prognostic factors for OS. DSG2 knockdown inhibited CC cell proliferation and migration. Conclusions DSG2 is a biomarker that promotes tumor proliferation and metastasis and is correlated with poor prognosis in early-stage CC.
Collapse
Affiliation(s)
- Shuhang Qin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Zhongshan Second Road 58, Guangzhou, 510080 People's Republic of China
| | - Yuandong Liao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Zhongshan Second Road 58, Guangzhou, 510080 People's Republic of China
| | - Qiqiao Du
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Zhongshan Second Road 58, Guangzhou, 510080 People's Republic of China
| | - Wei Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Zhongshan Second Road 58, Guangzhou, 510080 People's Republic of China
| | - Jiaming Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Zhongshan Second Road 58, Guangzhou, 510080 People's Republic of China
| | - Pan Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Zhongshan Second Road 58, Guangzhou, 510080 People's Republic of China
| | - Chunliang Shang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 People's Republic of China
| | - Tianyu Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Zhongshan Second Road 58, Guangzhou, 510080 People's Republic of China
| | - Meng Xia
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Zhongshan Second Road 58, Guangzhou, 510080 People's Republic of China
| | - Shuzhong Yao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Zhongshan Second Road 58, Guangzhou, 510080 People's Republic of China
| |
Collapse
|
9
|
Azimi A, Kaufman KL, Kim J, Ali M, Mann GJ, Fernandez-Penas P. Proteomics: An emerging approach for the diagnosis and classification of cutaneous squamous cell carcinoma and its precursors. J Dermatol Sci 2020; 99:9-16. [PMID: 32312638 DOI: 10.1016/j.jdermsci.2020.03.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 02/01/2020] [Accepted: 03/26/2020] [Indexed: 12/19/2022]
Abstract
Cutaneous squamous cell carcinoma (cSCC) and its precursors, actinic keratosis (AK) and Bowen's disease (BD), are the most common types of keratinocytic skin lesions (KSL) which account for the majority of non-melanoma skin cancer lethality. Currently, clinical and histopathological criteria are used for the diagnosis, classification and therapeutic intervention of KSLs, however discrepancies exist between the clinical presentations and histologic analyses of these lesions, making the diagnosis difficult. The identification of biomarkers as companion diagnostics for accurately stratifying KSL types is required to support the paradigm shift in current cancer care to personalised, precision medicine and ameliorate the negative impact of misdiagnoses or delayed diagnoses on patient outcome. Also, it is essential to elaborate on the poorly defined molecular modifications required for the initiation, development and progression of KSL from normal keratinocytes. By harnessing recent technological advances in molecular profiling techniques, it is anticipated that greater insight into the various combinations of proteomic events or alternative pathways underlying carcinogenesis will be gained. This review will explore recent genomic studies in KSL followed by assessing the feasibility and significance of mass spectrometry-based proteomics profiling as a promising approach to a better understanding of the oncogenic pathways underpinning the formation and progression of KSL lesions and in aiding the identification of novel biomarkers and new therapeutic targets. The development of non-invasive tools such as tape-stripping coupled with proteomic analysis alone or in conjunction with imaging and genomic technologies will complement existing clinical and histopathological parameters, leading to an improvement in patient outcomes.
Collapse
Affiliation(s)
- Ali Azimi
- Centre for Translational Skin Research, The University of Sydney, Westmead, Australia; Department of Dermatology, Westmead Hospital, Westmead, Australia
| | - Kimberley L Kaufman
- Department of Neurosurgery, Chris O'Brien Lifehouse, Camperdown, Australia; Discipline of Pathology, The University of Sydney, Camperdown, Australia
| | - Jennifer Kim
- Department of Tissue Pathology and Diagnostic Oncology, Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, Australia
| | - Marina Ali
- Centre for Translational Skin Research, The University of Sydney, Westmead, Australia; Department of Dermatology, Westmead Hospital, Westmead, Australia
| | - Graham J Mann
- Centre for Cancer Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, Australia; Melanoma Institute Australia, The University of Sydney, Wollstonecraft, Australia
| | - Pablo Fernandez-Penas
- Centre for Translational Skin Research, The University of Sydney, Westmead, Australia; Department of Dermatology, Westmead Hospital, Westmead, Australia.
| |
Collapse
|
10
|
Sun R, Ma C, Wang W, Yang S. Upregulation of desmoglein 2 and its clinical value in lung adenocarcinoma: a comprehensive analysis by multiple bioinformatics methods. PeerJ 2020; 8:e8420. [PMID: 32095325 PMCID: PMC7024574 DOI: 10.7717/peerj.8420] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 12/17/2019] [Indexed: 11/20/2022] Open
Abstract
Background Desmoglein-2 (DSG2), a desmosomal adhesion molecule, is found to be closely related to tumorigenesis in recent years. However, the clinical value of DSG2 in lung adenocarcinoma remains unclear. Methods Real-time reverse transcription-quantitative polymerase chain reaction (qRT-PCR) was utilized to detect the expression of DSG2 in 40 paired lung adenocarcinoma tissues and corresponding non-cancerous tissues. Data from The Cancer Genome Atlas (TCGA) and Oncomine datasets were also downloaded and analyzed. The correlation between DSG2 and clinicopathological features was investigated. The expression of DSG2 protein by immunohistochemical was also detected from tissue microarray and the Human Protein Atlas database. Integrated meta-analysis combining the three sources (qRT-PCR data, TCGA data and Oncomine datasets) was performed to evaluate the clinical value of DSG2. Univariate and multivariate Cox regression analyses were used to explore the prognostic value of DSG2. Then, co-expressed genes were calculated by Pearson correlation analysis. Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were used to investigate the underlying molecular mechanism. The expression level in lung adenocarcinoma and prognostic significance of the top ten co-expressed genes were searched from Gene Expression Profiling Interactive Analysis (GEPIA) online database. Results DSG2 was highly expressed in lung adenocarcinoma tissues based on qRT-PCR, TCGA and Oncomine datasets. The protein expression of DSG2 was also higher in lung adenocarcinoma. According to qRT-PCR and TCGA, high DSG2 expression was positively associated with tumor size (p = 0.027, p = 0.001), lymph node metastasis (p = 0.014, p < 0.001) and TNM stage (p = 0.023, p < 0.001). The combined standard mean difference values of DSG2 expression based on the three sources were 1.30 (95% confidence interval (CI): 1.08–1.52) using random effect model. The sensitivity and specificity were 0.73 (95% CI [0.69–0.76]) and 0.96 (95% CI [0.89–0.98]). The area under the curve based on summarized receiver operating characteristic (SROC) curve was 0.79 (95% CI [0.75–0.82]). Survival analysis revealed that high DSG2 expression was associated with a short overall survival (hazard ratio [HR] = 1.638; 95% CI [1.214–2.209], p = 0.001) and poor progression-free survival (HR = 1.475; 95% CI [1.102–1.974], p < 0.001). A total of 215 co-expressed genes were identified. According to GO and KEGG analyses, these co-expressed genes may be involved in “cell division”, “cytosol”, “ATP binding” and “cell cycle”. Based on GEPIA database, seven of the top ten co-expressed genes were highly expressed in lung adenocarcinoma (DSC2, SLC2A1, ARNTL2, ERO1L, ECT2, ANLN and LAMC2). High expression of these genes had shorter overall survival. Conclusions The expression of DSG2 is related to the tumor size, lymph node metastasis and TNM stage. Also, DSG2 predicts poor prognosis in lung adenocarcinoma.
Collapse
Affiliation(s)
- Ruiying Sun
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chao Ma
- Department of Anesthesiology, Xi'an Children Hospital, Xi'an, Shaanxi, China
| | - Wei Wang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Shuanying Yang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
11
|
Xie H, Xu H, Hou Y, Cai Y, Rong Z, Song W, Wang W, Li K. Integrative prognostic subtype discovery in high-grade serous ovarian cancer. J Cell Biochem 2019; 120:18659-18666. [PMID: 31347734 DOI: 10.1002/jcb.29049] [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: 04/02/2019] [Accepted: 04/30/2019] [Indexed: 01/19/2023]
Abstract
OBJECTIVE We sought to identify novel molecular subtypes of high-grade serous ovarian cancer (HGSC) by the integration of gene expression and proteomics data and to find the underlying biological characteristics of ovarian cancer to improve the clinical outcome. METHODS The iCluster method was utilized to analysis 131 common HGSC samples between TCGA and Clinical Proteomic Tumor Analysis Consortium databases. Kaplan-Meier survival curves were used to estimate the overall survival of patients, and the differences in survival curves were assessed using the log-rank test. RESULTS Two novel ovarian cancer subtypes with different overall survival (P = .00114) and different platinum status (P = .0061) were identified. Eighteen messenger RNAs and 38 proteins were selected as differential molecules between subtypes. Pathway analysis demonstrated arrhythmogenic right ventricular cardiomyopathy pathway played a critical role in the discrimination of these two subtypes and desmosomal cadherin DSG2, DSP, JUP, and PKP2 in this pathway were overexpression in subtype I compared with subtype II. CONCLUSION Our study extended the underlying prognosis-related biological characteristics of high-grade serous ovarian cancer. Enrichment of desmosomal cadherin increased the risk for HGSC prognosis among platinum-sensitive patients, the results guided the revision of the treatment options for platinum-sensitive ovarian cancer patients to improve outcomes.
Collapse
Affiliation(s)
- Hongyu Xie
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| | - Huan Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| | - Yan Hou
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| | - Yuqing Cai
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| | - Zhiwei Rong
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| | - Wei Song
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| | - Wenjie Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| | - Kang Li
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| |
Collapse
|
12
|
Vassal-Stermann E, Mottet M, Ducournau C, Iseni F, Vragniau C, Wang H, Zubieta C, Lieber A, Fender P. Mapping of Adenovirus of serotype 3 fibre interaction to desmoglein 2 revealed a novel 'non-classical' mechanism of viral receptor engagement. Sci Rep 2018; 8:8381. [PMID: 29849084 PMCID: PMC5976663 DOI: 10.1038/s41598-018-26871-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/18/2018] [Indexed: 12/05/2022] Open
Abstract
High-affinity binding of the trimeric fibre protein to a cell surface primary receptor is a common feature shared by all adenovirus serotypes. Recently, a long elusive species B adenovirus receptor has been identified. Desmoglein 2 (DSG2) a component of desmosomal junction, has been reported to interact at high affinity with Human adenoviruses HAd3, HAd7, HAd11 and HAd14. Little is known with respect to the molecular interactions of adenovirus fibre with the DSG2 ectodomain. By using different DSG2 ectodomain constructs and biochemical and biophysical experiments, we report that the third extracellular cadherin domain (EC3) of DSG2 is critical for HAd3 fibre binding. Unexpectedly, stoichiometry studies using multi-angle laser light scattering (MALLS) and analytical ultra-centrifugation (AUC) revealed a non-classical 1:1 interaction (one DSG2 per trimeric fibre), thus differentiating ‘DSG2-interacting’ adenoviruses from other protein receptor interacting adenoviruses in their infection strategy.
Collapse
Affiliation(s)
- Emilie Vassal-Stermann
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CNRS, CEA, 71 Avenue des Martyrs, 38042, Grenoble, France
| | - Manon Mottet
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CNRS, CEA, 71 Avenue des Martyrs, 38042, Grenoble, France
| | - Corinne Ducournau
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CNRS, CEA, 71 Avenue des Martyrs, 38042, Grenoble, France.,Unité de Virologie, Institut de Recherche Biomédicale des Armées, BP 73, 91223, Brétigny-sur-Orge Cedex, France
| | - Frédéric Iseni
- Unité de Virologie, Institut de Recherche Biomédicale des Armées, BP 73, 91223, Brétigny-sur-Orge Cedex, France
| | - Charles Vragniau
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CNRS, CEA, 71 Avenue des Martyrs, 38042, Grenoble, France
| | - Hongjie Wang
- University of Washington, Department of Medicine, Division of Medical Genetics, Box 357720, Seattle, WA, 98195, USA
| | - Chloe Zubieta
- Laboratoire de Physiologie Cellulaire et Végétale, Biosciences and Biotechnology Institute of Grenoble, UMR5168, CNRS/CEA/INRA/UGA, 17 Rue des Martyrs, 38054, Grenoble, France
| | - André Lieber
- University of Washington, Department of Medicine, Division of Medical Genetics, Box 357720, Seattle, WA, 98195, USA.
| | - Pascal Fender
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CNRS, CEA, 71 Avenue des Martyrs, 38042, Grenoble, France.
| |
Collapse
|
13
|
Regulation of CD44v6 expression in gastric carcinoma by the IL-6/STAT3 signaling pathway and its clinical significance. Oncotarget 2018; 8:45848-45861. [PMID: 28507278 PMCID: PMC5542232 DOI: 10.18632/oncotarget.17435] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 04/11/2017] [Indexed: 01/06/2023] Open
Abstract
As a cancer stem cell marker, CD44 variant 6 (CD44v6) has been implicated in carcinogenesis, tumor progression, and metastasis in a variety of human carcinomas. However, little is known about the expression of CD44v6 in Gastric Carcinoma (GC). Therefore we investigated CD44v6 expression in clinical specimen and further explore the underlying molecular mechanisms. In this study, we systemically investigated CD44v6 expression by immunohistochemistry in normal, premalignant gastric mucosa (low and high grade intraepithelial neoplasia), and GC at various stages. The correlation of CD44v6 expression with clinicopathological characteristics, and prognosis in GC was also analyzed. Next, we investigated cell proliferation, migration and invasion in GC cell lines. Furthermore, we explored a novel mechanism by which CD44V6 was upregulated in GC cell. The immunohistochemistry results showed that enhanced expression of CD44v6 was closely associated with tumor differentiation, lymph node metastasis, TNM stage and poor prognosis in GC patients. In gastric cancer cell lines, CD44v6 involved in cell proliferation, invasion and metastasis in Next, report on a novel mechanism by which interleukin-6/signal transducer and activator of transcription 3 (IL-6/STAT3) signaling up-regulates expression of CD44v6. RNA interference silencing of STAT3 resulted in decrease of CD44v6 levels. We also found that STAT3 inhibitor AG490 decrease expression of CD44v6 by blocking activation of STAT3, even in the presence of IL-6. Targeting STAT3-mediated CD44v6 up-regulation may represent a novel, effective treatment by eradicating the stomach tumor microenvironment.
Collapse
|
14
|
Overmiller AM, McGuinn KP, Roberts BJ, Cooper F, Brennan-Crispi DM, Deguchi T, Peltonen S, Wahl JK, Mahoney MG. c-Src/Cav1-dependent activation of the EGFR by Dsg2. Oncotarget 2018; 7:37536-37555. [PMID: 26918609 PMCID: PMC5122330 DOI: 10.18632/oncotarget.7675] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/25/2016] [Indexed: 12/31/2022] Open
Abstract
The desmosomal cadherin, desmoglein 2 (Dsg2), is deregulated in a variety of human cancers including those of the skin. When ectopically expressed in the epidermis of transgenic mice, Dsg2 activates multiple mitogenic signaling pathways and increases susceptibility to tumorigenesis. However, the molecular mechanism responsible for Dsg2-mediated cellular signaling is poorly understood. Here we show overexpression as well as co-localization of Dsg2 and EGFR in cutaneous SCCs in vivo. Using HaCaT keratinocytes, knockdown of Dsg2 decreases EGFR expression and abrogates the activation of EGFR, c-Src and Stat3, but not Erk1/2 or Akt, in response to EGF ligand stimulation. To determine whether Dsg2 mediates signaling through lipid microdomains, sucrose density fractionation illustrated that Dsg2 is recruited to and displaces Cav1, EGFR and c-Src from light density lipid raft fractions. STED imaging confirmed that the presence of Dsg2 disperses Cav1 from the cell-cell borders. Perturbation of lipid rafts with the cholesterol-chelating agent MβCD also shifts Cav1, c-Src and EGFR out of the rafts and activates signaling pathways. Functionally, overexpression of Dsg2 in human SCC A431 cells enhances EGFR activation and increases cell proliferation and migration through a c-Src and EGFR dependent manner. In summary, our data suggest that Dsg2 stimulates cell growth and migration by positively regulating EGFR level and signaling through a c-Src and Cav1-dependent mechanism using lipid rafts as signal modulatory platforms.
Collapse
Affiliation(s)
- Andrew M Overmiller
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kathleen P McGuinn
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Brett J Roberts
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA
| | - Felicia Cooper
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Donna M Brennan-Crispi
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Takahiro Deguchi
- Laboratory of Biophysics, Department of Cell Biology and Anatomy, University of Turku, Turku, Finland
| | - Sirkku Peltonen
- Department of Dermatology, University of Turku and Turku Hospital, Turku, Finland
| | - James K Wahl
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA
| | - Mỹ G Mahoney
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
15
|
Affiliation(s)
- Nicole A. Najor
- Department of Biology, University of Detroit Mercy, Detroit, Michigan 48221
| |
Collapse
|
16
|
Abstract
This research aims to explore the potential risk factors of lymph node metastasis (LNM) for early gastric cancers in young patients.We retrospectively collected data from 4287 patients who underwent gastrectomy from January 2005 to December 2015 at Linyi People's Hospital. Of these, we enrolled 397 eligible consecutive patients who had early gastric cancer, then divided them into 2 groups according to age (≤40 years and >40 years). The association between the clinicopathological factors and LNM was analyzed by univariate and multivariate analysis.Compared to older patients (>40 years), younger patients (≤40 years) with early gastric cancer had more diffuse and mixed types (51.1% and 37.8% vs 40% and 8.3%, respectively), less proximal gastric cancer (0% vs 33.8%, P < .01) and higher LNM (33.3% vs 13%, P < .01). Univariate analysis showed tumor invasion depth (P < .01), lymphovascular invasion (P < .01), and E-cadherin expression (P = .024) were associated with LNM in the younger cohort. Multivariate analysis revealed that lymphovascular invasion (OR = 17.740, 95% CI: 1.458-215.843) was an independent risk factor for LNM (P = .024). Further analysis showed 3 patients who were within expanded endoscopic resection indications were positive for LNM.Given the high risk of lymph node involvement in young patients with early gastric cancer, both endoscopic and surgical resection procedures should be performed with caution, and active postoperative surveillance is warranted.
Collapse
Affiliation(s)
- Tao Ji
- Department of Gastroenterology, Linyi People's Hospital, Linyi, Shandong Province
| | - Fan Zhou
- Department of Gastroenterology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - John Wang
- Department of Gastroenterology and Hepatology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Li Zi
- Department of General Surgery, Linyi People's Hospital, Linyi, Shandong Province, China
| |
Collapse
|
17
|
Zhou G, Yang L, Gray A, Srivastava AK, Li C, Zhang G, Cui T. The role of desmosomes in carcinogenesis. Onco Targets Ther 2017; 10:4059-4063. [PMID: 28860814 PMCID: PMC5565390 DOI: 10.2147/ott.s136367] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Desmosomes, which are intercellular adhesive complexes, are essential for the maintenance of epithelial homeostasis. They are located at the cell membrane, where they act as anchors for intermediate filaments. Downregulation of desmosome proteins in various cancers promotes tumor progression. However, the role of desmosomes in carcinogenesis is still being elucidated. Recent studies revealed that desmosome family members play a crucial role in tumor suppression or tumor promotion. This review focuses on studies that provide insights into the role of desmosomes in carcinogenesis and address their molecular functions.
Collapse
Affiliation(s)
- Guangxin Zhou
- Department of Oncology, Central Hospital of Binzhou, Binzhou Medical College, Binzhou, People's Republic of China
| | - Linlin Yang
- Department of Radiation Oncology, Arthur G James Hospital/Ohio State Comprehensive Cancer Center
| | | | - Amit Kumar Srivastava
- Division of Radiobiology, Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Gongwen Zhang
- Department of Cardiac Surgery, Central Hospital of Binzhou, Binzhou Medical College, Binzhou, People's Republic of China
| | - Tiantian Cui
- Department of Radiation Oncology, Arthur G James Hospital/Ohio State Comprehensive Cancer Center
| |
Collapse
|
18
|
Overmiller AM, Pierluissi JA, Wermuth PJ, Sauma S, Martinez-Outschoorn U, Tuluc M, Luginbuhl A, Curry J, Harshyne LA, Wahl JK, South AP, Mahoney MG. Desmoglein 2 modulates extracellular vesicle release from squamous cell carcinoma keratinocytes. FASEB J 2017; 31:3412-3424. [PMID: 28438789 DOI: 10.1096/fj.201601138rr] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 04/11/2017] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles (EVs) are nanoscale membrane-derived vesicles that serve as intercellular messengers carrying lipids, proteins, and genetic material. Substantial evidence has shown that cancer-derived EVs, secreted by tumor cells into the blood and other bodily fluids, play a critical role in modulating the tumor microenvironment and affecting the pathogenesis of cancer. Here we demonstrate for the first time that squamous cell carcinoma (SCC) EVs were enriched with the C-terminal fragment of desmoglein 2 (Dsg2), a desmosomal cadherin often overexpressed in malignancies. Overexpression of Dsg2 increased EV release and mitogenic content including epidermal growth factor receptor and c-Src. Inhibiting ectodomain shedding of Dsg2 with the matrix metalloproteinase inhibitor GM6001 resulted in accumulation of full-length Dsg2 in EVs and reduced EV release. When cocultured with Dsg2/green fluorescence protein-expressing SCC cells, green fluorescence protein signal was detected by fluorescence-activated cell sorting analysis in the CD90+ fibroblasts. Furthermore, SCC EVs activated Erk1/2 and Akt signaling and enhanced fibroblast cell proliferation. In vivo, Dsg2 was highly up-regulated in the head and neck SCCs, and EVs isolated from sera of patients with SCC were enriched in Dsg2 C-terminal fragment and epidermal growth factor receptor. This study defines a mechanism by which Dsg2 expression in cancer cells can modulate the tumor microenvironment, a step critical for tumor progression.-Overmiller, A. M., Pierluissi, J. A., Wermuth, P. J., Sauma, S., Martinez-Outschoorn, U., Tuluc, M., Luginbuhl, A., Curry, J., Harshyne, L. A., Wahl, J. K. III, South, A. P., Mahoney, M. G. Desmoglein 2 modulates extracellular vesicle release from squamous cell carcinoma keratinocytes.
Collapse
Affiliation(s)
- Andrew M Overmiller
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jennifer A Pierluissi
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Peter J Wermuth
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Sami Sauma
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Madalina Tuluc
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Adam Luginbuhl
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Joseph Curry
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Larry A Harshyne
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - James K Wahl
- Department of Oral Biology, University of Nebraska, Lincoln, Nebraska, USA
| | - Andrew P South
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Mỹ G Mahoney
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA;
| |
Collapse
|
19
|
Hütz K, Zeiler J, Sachs L, Ormanns S, Spindler V. Loss of desmoglein 2 promotes tumorigenic behavior in pancreatic cancer cells. Mol Carcinog 2017; 56:1884-1895. [PMID: 28277619 DOI: 10.1002/mc.22644] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 02/17/2017] [Accepted: 03/03/2017] [Indexed: 12/31/2022]
Abstract
The ability to maintain cell-cell adhesion is crucial for tissue integrity and organization. Accordingly, loss of cohesiveness plays a critical role in cancer invasion and metastasis. Desmosomes are cell junctions providing strong intercellular adhesive strength and dysregulation of desmosomal constituents contributes to cancer progression through altered cell signaling pathways. Here, we focused on the desmosomal adhesion molecules Desmoglein 2 (Dsg2) and Desmocollin 2 (Dsc2), and their contribution to migration and invasion in pancreatic cancer cells. Silencing of Dsg2 but not Dsc2 resulted in loss of cell cohesion and enhanced migration, and invasion of pancreatic adenocarcinoma cells. To identify potential pathways regulated by Dsg2, we performed kinase arrays and detected the activity of ERK and growth factor receptors to be significantly enhanced in Dsg2-deficient cells. Consequently, inhibition of ERK phosphorylation in Dsg2 knockdown cells normalized migration. Loss of Dsg2 resulted in reduced levels of the desmosomal adapter protein and transcriptional regulator Plakoglobin (PG) in an ERK-dependent manner, whereas other desmosomal molecules were not altered. Overexpression of PG rescued enhanced migration induced by silencing of Dsg2. These results identify a novel pro-migratory pathway of pancreatic cancer cells in which loss of Dsg2 reduces the levels of PG via deregulated MAPK signaling.
Collapse
Affiliation(s)
- Katharina Hütz
- Department I, Institute of Anatomy and Cell Biology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Julian Zeiler
- Department I, Institute of Anatomy and Cell Biology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Lena Sachs
- Department I, Institute of Anatomy and Cell Biology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Steffen Ormanns
- Institute of Pathology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Volker Spindler
- Department I, Institute of Anatomy and Cell Biology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| |
Collapse
|
20
|
Patel SJ, Darie CC, Clarkson BD. Effect of purified fractions from cell culture supernate of high-density pre-B acute lymphoblastic leukemia cells (ALL3) on the growth of ALL3 cells at low density. Electrophoresis 2016; 38:417-428. [PMID: 27804141 DOI: 10.1002/elps.201600399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/09/2016] [Accepted: 10/11/2016] [Indexed: 01/02/2023]
Abstract
The mechanisms underlying the aberrant growth and interactions between cells are not understood very well. The pre-B acute lymphoblastic leukemia cells directly obtained from an adult patient grow very poorly or do not grow at all at low density (LD), but grow better at high starting cell density (HD). We found that the LD ALL3 cells can be stimulated to grow in the presence of diffusible, soluble factors secreted by ALL3 cells themselves growing at high starting cell density. We then developed a biochemical purification procedure that allowed us to purify the factor(s) with stimulatory activity and analyzed them by nanoliquid chromatography-tandem mass spectrometry (nanoLC-MS/MS). Using nanoLC-MS/MS we have identified several proteins which were further processed using various bioinformatics tools. This resulted in eight protein candidates which might be responsible for the growth activity on non-growing LD ALL3 cells and their involvement in the stimulatory activity are discussed.
Collapse
Affiliation(s)
- Sapan J Patel
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology Program, New York, NY, USA.,Clarkson University, Biochemistry and Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY, USA
| | - Costel C Darie
- Clarkson University, Biochemistry and Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY, USA
| | - Bayard D Clarkson
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology Program, New York, NY, USA
| |
Collapse
|
21
|
Loiselle JJ, Roy JG, Sutherland LC. RBM5 reduces small cell lung cancer growth, increases cisplatin sensitivity and regulates key transformation-associated pathways. Heliyon 2016; 2:e00204. [PMID: 27957556 PMCID: PMC5133678 DOI: 10.1016/j.heliyon.2016.e00204] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/07/2016] [Accepted: 11/22/2016] [Indexed: 12/21/2022] Open
Abstract
Small cell lung cancer (SCLC) is the most aggressive type of lung cancer, with almost 95% of patients succumbing to the disease. Although RBM5, a tumor suppressor gene, is downregulated in the majority of lung cancers, its role in SCLC is unknown. Using the GLC20 SCLC cell line, which has a homozygous deletion encompassing the RBM5 gene locus, we established stable RBM5 expressing sublines and investigated the effects of RBM5 re-expression. Transcriptome and target identification studies determined that RBM5 directly regulates the cell cycle and apoptosis in SCLC cells, as well as significantly downregulates other important transformation-associated pathways such as angiogenesis and cell adhesion. RNA sequencing of paired non-tumor and tumor SCLC patient specimens showed decreased RBM5 expression in the tumors, and expression alterations in the majority of the same pathways that were altered in the GLC20 cells and sublines. Functional studies confirmed RBM5 expression slows SCLC cell line growth, and increases sensitivity to the chemotherapy drug cisplatin. Overall, our work demonstrates the importance of RBM5 expression to the non-transformed state of lung cells and the consequences of its deletion to SCLC development and progression.
Collapse
Affiliation(s)
- Julie J. Loiselle
- Biomolecular Sciences Program, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Justin G. Roy
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Leslie C. Sutherland
- Biomolecular Sciences Program, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Health Sciences North Research Institute (HSNRI), 41 Ramsey Lake Road, Sudbury, ON P3E 5J1, Canada
| |
Collapse
|
22
|
Celentano A, Mignogna MD, McCullough M, Cirillo N. Pathophysiology of the Desmo-Adhesome. J Cell Physiol 2016; 232:496-505. [PMID: 27505028 DOI: 10.1002/jcp.25515] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 08/08/2016] [Indexed: 01/18/2023]
Abstract
Advances in our understanding of desmosomal diseases have provided a clear demonstration of the key role played by desmosomes in tissue and organ physiology, highlighting the importance of their dynamic and finely regulated structure. In this context, non-desmosomal regulatory molecules have acquired increasing relevance in the study of this organelle resulting in extending the desmosomal interactome, named the "desmo-adhesome." Spatiotemporal changes in the expression and regulation of the desmo-adhesome underlie a number of genetic, infectious, autoimmune, and malignant conditions. The aim of the present article was to examine the structural and functional relationship of the desmosome, by providing a comprehensive, yet focused overview of the constituents targeted in human disease. The inclusion of the novel regulatory network in the desmo-adhesome pathophysiology opens new avenues to a deeper understanding of desmosomal diseases, potentially unveiling pathogenic mechanisms waiting to be explored. J. Cell. Physiol. 232: 496-505, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Antonio Celentano
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University Federico II of Naples, Naples, Italy.,Melbourne Dental School, University of Melbourne, Carlton, Victoria, Australia
| | - Michele Davide Mignogna
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University Federico II of Naples, Naples, Italy
| | - Michael McCullough
- Melbourne Dental School, University of Melbourne, Carlton, Victoria, Australia.,Oral Health Cooperative Research Centre (CRC), University of Melbourne, Carlton, Victoria, Australia
| | - Nicola Cirillo
- Melbourne Dental School, University of Melbourne, Carlton, Victoria, Australia.,Oral Health Cooperative Research Centre (CRC), University of Melbourne, Carlton, Victoria, Australia
| |
Collapse
|
23
|
Zhou F, Xu Y, Shi J, Lan X, Zou X, Wang L, Huang Q. Expression profile of E-cadherin, estrogen receptors, and P53 in early-onset gastric cancers. Cancer Med 2016; 5:3403-3411. [PMID: 27781410 PMCID: PMC5224840 DOI: 10.1002/cam4.931] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 08/26/2016] [Accepted: 09/04/2016] [Indexed: 12/12/2022] Open
Abstract
Early-onset gastric cancer (EOGC) is predominant in females, diffuse histology, and hereditary pattern. Germline mutation of CDH1 and p53 has been reported previously and female dominance was speculated to be associated with estrogen and its receptors. Expression of E-cadherin, estrogen receptor α (ERα), estrogen receptor β (ERβ), and p53 in EOGC remains unclear, which was the focus of this study, to assess clinical significance of their expression in EOGC. The expression of E-cadherin, ERα, ERβ, and p53 in tumors and normal tissues from surgically resected EOGCs was assessed by immunohistochemistry (n = 139) and Western blot (n = 7) methods, respectively. The expression in tumor tissues was significantly higher for ERα, ERβ, and p53, but lower for E-cadherin, compared to uninvolved mucosa. Positive staining of ERβ and p53 was more frequently observed in younger patients with advanced TNM stages. For E-cadherin, significant correlation was observed between the immunopositivity and TNM stages IA+IB. P53-negative patients had significantly better outcomes than p53-positive patients. Significant association between expression of E-cadherin and histologic types was found in familial, but not in sporadic, EOGC. In conclusion, our results demonstrated E-cadherin may have a role in initiation of EOGC and positive ERβ and p53 expression may partially explained early-onset and tumor progression of EOGC.
Collapse
Affiliation(s)
- Fan Zhou
- Department of Gastroenterology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yuanyuan Xu
- Department of Gastroenterology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jiong Shi
- Department of Pathology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xing Lan
- Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaoping Zou
- Department of Gastroenterology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Lei Wang
- Department of Gastroenterology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Qin Huang
- Department of Pathology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China.,Department of Pathology, VA Boston Healthcare System and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
24
|
Abstract
Alzheimer's disease (AD) is a progressive, neurodegenerative disease and the most common form of dementia in elderly people. It is an emerging public health problem that poses a huge societal burden. Linkage analysis was the first milestone in unraveling the mutations in APP, PSEN1, and PSEN2 that cause early-onset AD, followed by the discovery of apolipoprotein E-ε4 allele as the only one genetic risk factor for late-onset AD. Genome-wide association studies have revolutionized genetic research and have identified over 20 genetic loci associated with late-onset AD. Recently, next-generation sequencing technologies have enabled the identification of rare disease variants, including unmasking small mutations with intermediate risk of AD in PLD3, TREM2, UNC5C, AKAP9, and ADAM10. This review provides an overview of the genetic basis of AD and the relationship between these risk genes and the neuropathologic features of AD. An understanding of genetic mechanisms underlying AD pathogenesis and the potentially implicated pathways will lead to the development of novel treatment for this devastating disease.
Collapse
Affiliation(s)
- Mohan Giri
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, People’s Republic of China
| | - Man Zhang
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, People’s Republic of China
| | - Yang Lü
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, People’s Republic of China
| |
Collapse
|
25
|
Ormanns S, Altendorf-Hofmann A, Jackstadt R, Horst D, Assmann G, Zhao Y, Bruns C, Kirchner T, Knösel T. Desmogleins as prognostic biomarkers in resected pancreatic ductal adenocarcinoma. Br J Cancer 2015; 113:1460-6. [PMID: 26469831 PMCID: PMC4815888 DOI: 10.1038/bjc.2015.362] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 09/10/2015] [Accepted: 09/18/2015] [Indexed: 12/21/2022] Open
Abstract
Background: Frequent disease relapse and a lack of effective therapies result in a very poor outcome in pancreatic ductal adenocarcinoma (PDAC) patients. Thus, identification of prognostic biomarkers and possible therapeutic targets is essential. Besides their function in cell–cell adhesion, desmogleins may play a role in tumour progression and invasion that has not been investigated in PDAC to date. This study evaluated desmoglein expression as a biomarker in PDAC. Methods: Using immunohistochemistry, we examined desmoglein 1 (DSG1), desmoglein 2 (DSG2) and desmoglein 3 (DSG3) expression in the tumour tissue of 165 resected PDAC cases. Expression levels were correlated to the patients' clinicopathological parameters and postoperative survival times. We confirmed these results in two independent gene expression data sets. Results: A total of 36% of the tumours showed high DSG3 expression that correlated significantly with shorter patient survival (P=0.011) and poor tumour differentiation (P<0.001), whereas no such association was detected for DSG1 or DSG2. In RNA-Seq data and in microarray expression data, high DSG3 expression correlated significantly with poor survival (P=0.000356 and P=0.00499). Conclusions: We identify DSG3 as a negative prognostic biomarker in resected PDAC, as high DSG3 expression is associated with poor overall survival and poor tumour-specific survival. These findings suggest DSG3 and its downstream signalling pathways as possible therapeutic targets in DSG3-expressing PDAC.
Collapse
Affiliation(s)
- Steffen Ormanns
- Institute of Pathology, Ludwig-Maximilians-University, Thalkirchner Strasse 36, Munich 80337, Germany
| | - Annelore Altendorf-Hofmann
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Erlanger Allee 101, Jena 07747, Germany
| | - Rene Jackstadt
- Cancer Research UK, Beatson Institute, Glasgow G61 1BD, UK
| | - David Horst
- Institute of Pathology, Ludwig-Maximilians-University, Thalkirchner Strasse 36, Munich 80337, Germany
| | - Gerald Assmann
- Institute of Pathology, Ludwig-Maximilians-University, Thalkirchner Strasse 36, Munich 80337, Germany
| | - Yue Zhao
- Department of Surgery, University Hospital Magdeburg, Leipziger Strasse 44, Magdeburg 39120, Germany
| | - Christiane Bruns
- Department of Surgery, University Hospital Magdeburg, Leipziger Strasse 44, Magdeburg 39120, Germany
| | - Thomas Kirchner
- Institute of Pathology, Ludwig-Maximilians-University, Thalkirchner Strasse 36, Munich 80337, Germany
| | - Thomas Knösel
- Institute of Pathology, Ludwig-Maximilians-University, Thalkirchner Strasse 36, Munich 80337, Germany
| |
Collapse
|
26
|
Abstract
Desmosomes represent adhesive, spot-like intercellular junctions that in association with intermediate filaments mechanically link neighboring cells and stabilize tissue architecture. In addition to this structural function, desmosomes also act as signaling platforms involved in the regulation of cell proliferation, differentiation, migration, morphogenesis, and apoptosis. Thus, deregulation of desmosomal proteins has to be considered to contribute to tumorigenesis. Proteolytic fragmentation and downregulation of desmosomal cadherins and plaque proteins by transcriptional or epigenetic mechanisms were observed in different cancer entities suggesting a tumor-suppressive role. However, discrepant data in the literature indicate that context-dependent differences based on alternative intracellular, signal transduction lead to altered outcome. Here, modulation of Wnt/β-catenin signaling by plakoglobin or desmoplakin and of epidermal growth factor receptor signaling appears to be of special relevance. This review summarizes current evidence on how desmosomal proteins participate in carcinogenesis, and depicts the molecular mechanisms involved.
Collapse
Affiliation(s)
- Otmar Huber
- a Institute of Biochemistry II, Jena University Hospital, Friedrich-Schiller-University Jena , Nonnenplan 2-4, 07743 Jena , Germany.,b Center for Sepsis Control and Care, Jena University Hospital , Erlanger Allee 101, 07747 Jena , Germany
| | - Iver Petersen
- c Institute of Pathology, Jena University Hospital, Friedrich-Schiller-University Jena , Ziegelmühlenweg 1, 07743 Jena , Germany
| |
Collapse
|
27
|
Mehrian-Shai R, Freedman S, Shams S, Doherty J, Slattery W, Hsu NYH, Reichardt JKV, Andalibi A, Toren A. Schwannomas exhibit distinct size-dependent gene-expression patterns. Future Oncol 2015; 11:1751-8. [PMID: 26075443 DOI: 10.2217/fon.15.72] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Neurofibromatosis type 2 (NF2)-associated vestibular schwannomas have variable size at presentation which presents a unique challenge in NF2 patient management. Therefore, we investigated the molecular signature characteristic of the differences in size for improved individualized precise therapy. MATERIALS & METHODS RNA expression analysis was performed on 15 small and 27 large NF2-associated vestibular schwannoma tumors using a microarray analyzing over 47,000 transcripts. RESULTS A signature of 11 genes was found to be correlated with NF2 tumor size. CONCLUSION We have identified the genetic hallmark that differentiates large NF2-associated tumors from smaller tumors. This is the first time that these genes have been shown to be the hallmark for NF2 tumor size.
Collapse
Affiliation(s)
- Ruty Mehrian-Shai
- Department of Pediatric Hemato-Oncology, The Cancer Research Center, Sheba Medical Center, 2 Sheba Road, Ramat Gan, 52621, Israel
| | - Shany Freedman
- Department of Pediatric Hemato-Oncology, The Cancer Research Center, Sheba Medical Center, 2 Sheba Road, Ramat Gan, 52621, Israel
| | - Soheil Shams
- BioDiscovery, 5155 W Rosecrans Ave # 310, Hawthorne, CA 90250, USA
| | - Joni Doherty
- Head & Neck Surgery, University of California, San Diego School of Medicine, 9500 Gilman Dr, La Jolla, CA 92093, USA
| | - William Slattery
- Department of Clinical Studies, House Ear Institute, 2100 W 3rd St #500, Los Angeles, CA 90057, USA
| | | | - Juergen K V Reichardt
- Division of Tropical Health & Medicine, James Cook University, Townsville, QLD, Australia
| | - Ali Andalibi
- Stony Brook University, Stony Brook, NY 11794, USA
| | - Amos Toren
- Department of Pediatric Hemato-Oncology, The Cancer Research Center, Sheba Medical Center, 2 Sheba Road, Ramat Gan, 52621, Israel
| |
Collapse
|
28
|
Abstract
Desmosomes are cell-cell junctions that mediate adhesion and couple the intermediate filament cytoskeleton to sites of cell-cell contact. This architectural arrangement integrates adhesion and cytoskeletal elements of adjacent cells. The importance of this robust adhesion system is evident in numerous human diseases, both inherited and acquired, which occur when desmosome function is compromised. This review focuses on autoimmune and infectious diseases that impair desmosome function. In addition, we discuss emerging evidence that desmosomal genes are often misregulated in cancer. The emphasis of our discussion is placed on the way in which human diseases can inform our understanding of basic desmosome biology and in turn, the means by which fundamental advances in the cell biology of desmosomes might lead to new treatments for acquired diseases of the desmosome.
Collapse
|
29
|
Gupta A, Nitoiu D, Brennan-Crispi D, Addya S, Riobo NA, Kelsell DP, Mahoney MG. Cell cycle- and cancer-associated gene networks activated by Dsg2: evidence of cystatin A deregulation and a potential role in cell-cell adhesion. PLoS One 2015; 10:e0120091. [PMID: 25785582 PMCID: PMC4364902 DOI: 10.1371/journal.pone.0120091] [Citation(s) in RCA: 19] [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: 09/05/2014] [Accepted: 02/02/2015] [Indexed: 01/06/2023] Open
Abstract
Cell-cell adhesion is paramount in providing and maintaining multicellular structure and signal transmission between cells. In the skin, disruption to desmosomal regulated intercellular connectivity may lead to disorders of keratinization and hyperproliferative disease including cancer. Recently we showed transgenic mice overexpressing desmoglein 2 (Dsg2) in the epidermis develop hyperplasia. Following microarray and gene network analysis, we demonstrate that Dsg2 caused a profound change in the transcriptome of keratinocytes in vivo and altered a number of genes important in epithelial dysplasia including: calcium-binding proteins (S100A8 and S100A9), members of the cyclin protein family, and the cysteine protease inhibitor cystatin A (CSTA). CSTA is deregulated in several skin cancers, including squamous cell carcinomas (SCC) and loss of function mutations lead to recessive skin fragility disorders. The microarray results were confirmed by qPCR, immunoblotting, and immunohistochemistry. CSTA was detected at high level throughout the newborn mouse epidermis but dramatically decreased with development and was detected predominantly in the differentiated layers. In human keratinocytes, knockdown of Dsg2 by siRNA or shRNA reduced CSTA expression. Furthermore, siRNA knockdown of CSTA resulted in cytoplasmic localization of Dsg2, perturbed cytokeratin 14 staining and reduced levels of desmoplakin in response to mechanical stretching. Both knockdown of either Dsg2 or CSTA induced loss of cell adhesion in a dispase-based assay and the effect was synergistic. Our findings here offer a novel pathway of CSTA regulation involving Dsg2 and a potential crosstalk between Dsg2 and CSTA that modulates cell adhesion. These results further support the recent human genetic findings that loss of function mutations in the CSTA gene result in skin fragility due to impaired cell-cell adhesion: autosomal-recessive exfoliative ichthyosis or acral peeling skin syndrome.
Collapse
Affiliation(s)
- Abhilasha Gupta
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Daniela Nitoiu
- Center for Cutaneous Research, Blizard Institute, Barts and the London School or Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Donna Brennan-Crispi
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Sankar Addya
- Kimmel Cancer Center, Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Natalia A. Riobo
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - David P. Kelsell
- Center for Cutaneous Research, Blizard Institute, Barts and the London School or Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Mỹ G. Mahoney
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
30
|
Broussard JA, Getsios S, Green KJ. Desmosome regulation and signaling in disease. Cell Tissue Res 2015; 360:501-12. [PMID: 25693896 DOI: 10.1007/s00441-015-2136-5] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 01/21/2015] [Indexed: 01/10/2023]
Abstract
Desmosomes are cell-cell adhesive organelles with a well-known role in forming strong intercellular adhesion during embryogenesis and in adult tissues subject to mechanical stress, such as the heart and skin. More recently, desmosome components have also emerged as cell signaling regulators. Loss of expression or interference with the function of desmosome molecules results in diseases of the heart and skin and contributes to cancer progression. However, the underlying molecular mechanisms that result in inherited and acquired disorders remain poorly understood. To address this question, researchers are directing their studies towards determining the functions that occur inside and outside of the junctions and the extent to which functions are adhesion-dependent or independent. This review focuses on recent discoveries that provide insights into the role of desmosomes and desmosome components in cell signaling and disease; wherever possible, we address molecular functions within and outside of the adhesive structure.
Collapse
Affiliation(s)
- Joshua A Broussard
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | | | | |
Collapse
|
31
|
Pietkiewicz P, Gornowicz-Porowska J, Bowszyc-Dmochowska M, Jagielska J, Helak-Łapaj C, Kaczmarek E, Dmochowski M. Discordant expression of desmoglein 2 and 3 at the mRNA and protein levels in nodular and superficial basal cell carcinoma revealed by immunohistochemistry and fluorescent in situ hybridization. Clin Exp Dermatol 2015; 39:628-35. [PMID: 24934917 DOI: 10.1111/ced.12355] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2013] [Indexed: 12/17/2022]
Abstract
BACKGROUND Basal cell carcinoma (BCC) is the most common human cancer. It is thought that skewed expression of desmogleins (Dsgs) in BCC may promote tumourigenesis. AIM To comparatively examine expression of Dsg2/Dsg3, using fluorescent in situ hybridization (FISH) and immunohistochemistry (IHC) in BCC subtypes. METHODS In total, 84 frozen sections from patients with various clinical or histological subtypes of BCC were analyzed. Expressions of Dsg2/Dsg3 protein and Dsg2/Dsg3 mRNA were evaluated using IHC and FISH, respectively, in BCC nests and BCC-free epidermis, and then quantitatively measured. RESULTS There was loss of correlation between Dsg2 and Dsg3 (IHC) in nodular and superficial BCC (nBCC, sBCC), and significant correlation between Dsg2 and Dsg3 (FISH) in BCC, but not nBCC and sBCC. CONCLUSIONS Because more prominent aberrations of Dsg2/Dsg3 expression were seen at the protein than at the mRNA level in BCC, these comparative observations indicate greater importance of events at the proteome level than those at the genome level in tumour functional compartments. Different Dsg2/Dsg3 expression in sBCC and nBCC might corroborate the possibility that sBCC and nBCC are separate conditions. These results may contribute to better understanding of the biological behaviour of BCC.
Collapse
Affiliation(s)
- P Pietkiewicz
- Autoimmune Blistering Dermatoses Section, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | | | | | | | | |
Collapse
|
32
|
Brown L, Wan H. Desmoglein 3: a help or a hindrance in cancer progression? Cancers (Basel) 2015; 7:266-86. [PMID: 25629808 PMCID: PMC4381258 DOI: 10.3390/cancers7010266] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/09/2015] [Accepted: 01/16/2015] [Indexed: 02/07/2023] Open
Abstract
Desmoglein 3 is one of seven desmosomal cadherins that mediate cell-cell adhesion in desmosomes. Desmosomes are the intercellular junctional complexes that anchor the intermediate filaments of adjacent cells and confer strong cell adhesion thus are essential in the maintenance of tissue architecture and structural integrity. Like adherens junctions, desmosomes function as tumour suppressors and are down regulated in the process of epithelial-mesenchymal transition and in tumour cell invasion and metastasis. However, recently several studies have shown that various desmosomal components, including desmoglein 3, are up-regulated in cancer with increased levels of expression correlating with the clinical stage of malignancy, implicating their potentiality to serve as a diagnostic and prognostic marker. Furthermore, in vitro studies have demonstrated that overexpression of desmoglein 3 in cancer cell lines activates several signal pathways that have an impact on cell morphology, adhesion and locomotion. These additional signalling roles of desmoglein 3 may not be associated to its adhesive function in desmosomes but rather function outside of the junctions, acting as a key regulator in the control of actin based cellular processes. This review will discuss recent advances which support the role of desmoglein 3 in cancer progression.
Collapse
Affiliation(s)
- Louise Brown
- Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Center for Clinical and Diagnostic Oral Sciences, Institute of Dentistry, Blizard Building, London E1 2AT, UK.
| | - Hong Wan
- Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Center for Clinical and Diagnostic Oral Sciences, Institute of Dentistry, Blizard Building, London E1 2AT, UK.
| |
Collapse
|
33
|
O'Shea C, Fitzpatrick JE, Koch PJ. Desmosomal defects in acantholytic squamous cell carcinomas. J Cutan Pathol 2014; 41:873-9. [PMID: 25264142 DOI: 10.1111/cup.12390] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 07/11/2014] [Accepted: 07/20/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND Acantholytic squamous cell carcinoma (Acantholytic SCC) are epithelial tumors characterized by a loss of cell adhesion between neoplastic keratinocytes. The mechanism underlying loss of cell-cell adhesion in these tumors is not understood. METHODS A retrospective analysis of acantholytic SCC (n = 17) and conventional SCC (n = 16, controls not showing acantholysis) was conducted using a set of desmosomal and adherens junction protein antibodies. Immunofluorescence microscopy was used to identify tumors with loss of adhesion protein expression. RESULTS The vast majority of acantholytic SCC (89%) showed focal loss of at least one desmosomal cell adhesion protein. Most interestingly, 65% of these tumors lost expression of two or more desmosomal proteins. CONCLUSIONS Loss of cell adhesion in acantholytic SCC is most likely linked to the focal loss of desmosomal protein expression, thus providing potential mechanistic insight into the patho-mechanism underlying this malignancy.
Collapse
Affiliation(s)
- Charlene O'Shea
- Department of Dermatology, University of Colorado School of Medicine, Aurora, CO, USA; Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | | | | |
Collapse
|
34
|
Riss D, Pammer J, Grasl MC, Kaider A, Schneider S, Erovic BM. No evidence for desmocollin 3 to serve as a prognostic marker in primary radiotherapy of head and neck cancer. Wien Klin Wochenschr 2014; 127:24-30. [PMID: 25301099 DOI: 10.1007/s00508-014-0618-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 07/31/2014] [Indexed: 11/27/2022]
Abstract
PURPOSE To investigate the role of desmocollin 3 expression in radiation sensitivity and survival in squamous cell carcinoma of the head and neck. METHODS Tissue samples of 40 patients undergoing primary radiotherapy were stained for desmocollin 3, a transmembrane desmosomal protein. The protein expression was determined by immunohistochemistry. A retrospective chart review was performed to assess overall survival, radiation sensitivity, and disease-free interval. RESULTS 18 of 40 patients showed desmocollin 3 immunoreactivity. 78 % (14/18) showed radiation sensitivity in the desmocollin 3 positive group, whereas 59 % (13/22) underwent remission in the desmocollin 3 negative group (p = 0.2). The median overall and disease-free survival time was 21 and 26 months, respectively. Univariate and multivariable Cox regression models revealed no statistically significant hazard ratios. CONCLUSION In this cohort of squamous cell carcinoma of the head and neck, we found no evidence for a predictive potential of desmocollin 3.
Collapse
Affiliation(s)
- Dominik Riss
- Department of Otolaryngology, Head and Neck Surgery, Medical University of Vienna, Waehriger Guertel 18-20, 1090, Vienna, Austria,
| | | | | | | | | | | |
Collapse
|
35
|
Barber AG, Castillo-Martin M, Bonal DM, Rybicki BA, Christiano AM, Cordon-Cardo C. Characterization of desmoglein expression in the normal prostatic gland. Desmoglein 2 is an independent prognostic factor for aggressive prostate cancer. PLoS One 2014; 9:e98786. [PMID: 24896103 PMCID: PMC4045811 DOI: 10.1371/journal.pone.0098786] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 05/07/2014] [Indexed: 11/18/2022] Open
Abstract
PURPOSE The expression of desmogleins (DSGs), which are known to be crucial for establishing and maintaining the cell-cell adhesion required for tissue integrity, has been well characterized in the epidermis and hair follicle; however, their expression in other epithelial tissues such as prostate is poorly understood. Although downregulation of classical cadherins, such as E-cadherin, has been described in prostate cancer tissue samples, the expression of desmogleins has only been previously reported in prostate cancer cell lines. In this study we characterized desmoglein expression in normal prostate tissues, and further investigated whether Desmoglein 2 (DSG2) expression specifically can serve as a potential clinical prognostic factor for patients diagnosed with primary prostate cancer. EXPERIMENTAL DESIGN We utilized immunofluorescence to examine DSG2 expression in normal prostate (n = 50) and in a clinically well-characterized cohort of prostate cancer patients (n = 414). Correlation of DSG2 expression with clinico-pathological characteristics and biochemical recurrence was analyzed to assess its clinical significance. RESULTS These studies revealed that DSG2 and DSG4 were specifically expressed in prostatic luminal cells, whereas basal cells lack their expression. In contrast, DSG1 and DSG3 were not expressed in normal prostate epithelium. Further analyses of DSG2 expression in prostate cancer revealed that reduced levels of this biomarker were a significant independent marker of poor clinical outcome. CONCLUSION Here we report for the first time that a low DSG2 expression phenotype is a useful prognostic biomarker of tumor aggressiveness and may serve as an aid in identifying patients with clinically significant prostate cancer.
Collapse
Affiliation(s)
- Alison G. Barber
- Department of Genetics and Development, Columbia University, New York, New York, United States of America
| | - Mireia Castillo-Martin
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- * E-mail: (MCM); (CCC)
| | - Dennis M. Bonal
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Benjamin A. Rybicki
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Angela M. Christiano
- Department of Genetics and Development, Columbia University, New York, New York, United States of America
- Department of Dermatology, Columbia University, New York, New York, United States of America
| | - Carlos Cordon-Cardo
- Department of Pathology and Cell Biology, Columbia University, New York, New York, United States of America
- Department of Urology, Columbia University, New York, New York, United States of America
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York, United States of America
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- * E-mail: (MCM); (CCC)
| |
Collapse
|
36
|
Fang WK, Chen B, Xu XE, Liao LD, Wu ZY, Wu JY, Shen J, Xu LY, Li EM. Altered expression and localization of desmoglein 3 in esophageal squamous cell carcinoma. Acta Histochem 2014; 116:803-9. [PMID: 24630396 DOI: 10.1016/j.acthis.2014.01.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 01/14/2014] [Accepted: 01/16/2014] [Indexed: 02/05/2023]
Abstract
Desmoglein 3 (DSG3), a transmembrane cadherin of the desmosomal cell-cell adhesion structure, plays vital roles in the maintenance of normal epithelial tissue architecture. Reports implicating a role for DSG3 expression in cancer are few and contradictory. In this study, immunohistochemical staining was employed to investigate DSG3 expression and subcellular localization in esophageal squamous cell carcinoma (ESCC), and to correlate changes with clinical characteristics. Results indicate that in normal squamous cell epithelia, strong DSG3 immunoreactivity was observed in the Stratum spinosum, and localization occurred only at the cell membrane. In ESCC, DSG3 immunoreactivity displayed an abnormal cytoplasmic localization that was correlated with cell differentiation (P=0.018). Most strikingly, in 74.1% of the tumors, DSG3 expression was up-regulated and correlated with regional lymph node metastasis (P=0.036). Moreover, in patients without lymph node metastasis, cytoplasmic localization of DSG3 correlated with poor prognosis (P=0.044). These results suggest that DSG3 is involved in the development of ESCC and imply that DSG3 overexpression is likely to be an essential contributor to the aggressive features of esophageal cancer.
Collapse
Affiliation(s)
- Wang-Kai Fang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China; The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
| | - Bo Chen
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, China
| | - Xiu-E Xu
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, China
| | - Lian-Di Liao
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, China
| | - Zhi-Yong Wu
- Department of Oncology Surgery, Affiliated Shantou Hospital of Sun Yat-Sen University, Shantou, China
| | - Jian-Yi Wu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
| | - Jian Shen
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, China
| | - Li-Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou, China.
| | - En-Min Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China; The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China.
| |
Collapse
|
37
|
Aberrant expression and altered cellular localization of desmosomal and hemidesmosomal proteins are associated with aggressive clinicopathological features of oral squamous cell carcinoma. Virchows Arch 2014; 465:35-47. [PMID: 24849508 DOI: 10.1007/s00428-014-1594-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 04/22/2014] [Accepted: 05/12/2014] [Indexed: 10/25/2022]
Abstract
Disruption of cell adhesion plays a central role in dedifferentiation, invasion, and metastasis of various cancers. The desmosome and hemidesmosome are anchoring junctions that control cell-cell and cell-matrix adhesion, respectively. To clarify their contributions in mediating the biological properties of oral cancer, we immunohistochemically examined the expression of desmoglein 1 (DSG1), DSG2, DSG3, desmocollin 2 (DSC2), integrin beta 4 (ITGB4), laminin gamma chain 2 (LAMC2), and collagen type 17 alpha 1 (COL17A1) in 51 cases of oral squamous cell carcinoma. On normal oral epithelial cells, DSG1, DSG3, DSC2, and COL17A1 were expressed on the plasma membrane, while ITGB4 and mature LAMC2 were present at the basement membrane. In cancer, the expression of DSG1, DSG3, DSC2, and COL17A1 decreased and internalized to the cytoplasm. Cytoplasmic expression of DSG2, ITGB4, and LAMC2 was induced in the cancer cells facing to the stroma. We scored immunohistochemical expression and correlated this to clinicopathological parameters including histologic differentiation, pattern of invasion, and presence of lymph node metastasis. Decrease of DSG3 and DSC2 expression correlated with a more aggressive cancer phenotype: less differentiated and more invasive histologic features and a higher incidence of nodal metastasis. Lower COL17A1 and higher LAMC2 expression were also associated with a more aggressive phenotype. The present study demonstrates that aberrant expression and altered cellular localization of desmosomal and hemidesmosomal proteins are associated with aggressive clinicopathological features of oral cancer. This reinforces the notion that disturbance of the keratin-associated anchoring junctions confers aggressive features to cancer cells.
Collapse
|
38
|
Peitsch WK, Doerflinger Y, Fischer-Colbrie R, Huck V, Bauer AT, Utikal J, Goerdt S, Schneider SW. Desmoglein 2 depletion leads to increased migration and upregulation of the chemoattractant secretoneurin in melanoma cells. PLoS One 2014; 9:e89491. [PMID: 24558503 PMCID: PMC3928442 DOI: 10.1371/journal.pone.0089491] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 01/23/2014] [Indexed: 12/21/2022] Open
Abstract
During development and progression of malignant melanoma, an important role has been attributed to alterations of cell-cell adhesions, in particular, to a “cadherin switch” from E- to N-cadherin. We have previously shown that a subtype of melanoma cells express the desmosomal cadherin desmoglein 2 as non-junction-bound cell surface protein in addition to classical cadherins. To study the role of desmoglein 2 in melanoma cells, melanoma lines containing high endogenous amounts of desmoglein 2 were depleted of the protein by RNA interference. Transwell migration and scratch wounding assays showed markedly increased migration upon desmoglein 2 suppression whereas proliferation and viability remained unaltered. In gene expression profiles, desmoglein 2 depletion was associated with overexpression of migration-related genes. Strongest overexpression was found for secretogranin II which has not been reported in melanoma cells before. The bioactive peptide derived from secretogranin II, secretoneurin, is known to exert chemoattractive functions and was demonstrated here to stimulate melanoma cell migration. In summary, we show that desmoglein 2 expression attenuates migration of melanoma cells. The mechanism of desmoglein 2 impaired cell migration is mediated by downregulation of secretogranin II. Loss of desmoglein 2 increases expression of secretogranin II, followed by an enhanced migratory activity of melanoma cells. Our data add a new pathway of regulating melanoma cell migration related to a desmoglein 2 – secretogranin II axis.
Collapse
Affiliation(s)
- Wiebke K. Peitsch
- Department of Dermatology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Helmholtz Group for Cell Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- * E-mail:
| | - Yvette Doerflinger
- Department of Dermatology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Helmholtz Group for Cell Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Volker Huck
- Department of Dermatology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexander T. Bauer
- Department of Dermatology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Jochen Utikal
- Department of Dermatology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sergij Goerdt
- Department of Dermatology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan W. Schneider
- Department of Dermatology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
39
|
|
40
|
Fang WK, Gu W, Liao LD, Chen B, Wu ZY, Wu JY, Shen J, Xu LY, Li EM. Prognostic significance of desmoglein 2 and desmoglein 3 in esophageal squamous cell carcinoma. Asian Pac J Cancer Prev 2014; 15:871-6. [PMID: 24568510 DOI: 10.7314/apjcp.2014.15.2.871] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Desmogleins (DSGs) are major members among the desmosomal cadherins critically involved in cell-cell adhesion and the maintenance of normal tissue architecture in epithelia. Reports exploring links of DSG family member expression with cancers are few and vary. The aim of this study was to investigate the ratio of DSG2 and DSG3 mRNA expression in esophageal squamous cell carcinoma (ESCC) tissue to normal tissue (T/N ratio) and evaluate correlations with clinical parameters. METHODS The mRNA expression of DSGs, as well as γ-catenin and desmoplakin, was detected by real-time quantitative RT-PCR in 85 cases of ESCC tissue specimens. RESULTS The expression level of DSG3 mRNA was significantly higher than that of DSG2 in ESCC specimens (p = 0.000). DSG3 mRNA expression highly correlated with histological grade (p = 0.009), whereas that of DSG2 did not significantly relate to any clinicopathologic parameter. Kaplan-Meier survival analysis showed that only DSG3 expression had an impact on the survival curve, with negative DSG3 expression indicating worse survival (p = 0.038). Multivariate Cox regression analysis demonstrated DSG3 to be an independent prognostic factor for survival. Furthermore, correlation analysis demonstrated the mRNA level of DSG3 to highly correlate with those of γ-catenin and desmoplakin in ESCC samples (p=0.000), implying that the expression of desmosomal components might be regulated by the same upstream regulatory molecules. CONCLUSIONS Our findings suggest that DSG3 may be involved in the progression of ESCC and serve as a prognostic marker, while expression of DSG2 cannot be used as a predictor of ESCC patient outcome.
Collapse
Affiliation(s)
- Wang-Kai Fang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China E-mail : ,
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Al-Jassar C, Bikker H, Overduin M, Chidgey M. Mechanistic basis of desmosome-targeted diseases. J Mol Biol 2013; 425:4006-22. [PMID: 23911551 PMCID: PMC3807649 DOI: 10.1016/j.jmb.2013.07.035] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 07/23/2013] [Accepted: 07/24/2013] [Indexed: 11/21/2022]
Abstract
Desmosomes are dynamic junctions between cells that maintain the structural integrity of skin and heart tissues by withstanding shear forces. Mutations in component genes cause life-threatening conditions including arrhythmogenic right ventricular cardiomyopathy, and desmosomal proteins are targeted by pathogenic autoantibodies in skin blistering diseases such as pemphigus. Here, we review a set of newly discovered pathogenic alterations and discuss the structural repercussions of debilitating mutations on desmosomal proteins. The architectures of native desmosomal assemblies have been visualized by cryo-electron microscopy and cryo-electron tomography, and the network of protein domain interactions is becoming apparent. Plakophilin and desmoplakin mutations have been discovered to alter binding interfaces, structures, and stabilities of folded domains that have been resolved by X-ray crystallography and NMR spectroscopy. The flexibility within desmoplakin has been revealed by small-angle X-ray scattering and fluorescence assays, explaining how mechanical stresses are accommodated. These studies have shown that the structural and functional consequences of desmosomal mutations can now begin to be understood at multiple levels of spatial and temporal resolution. This review discusses the recent structural insights and raises the possibility of using modeling for mechanism-based diagnosis of how deleterious mutations alter the integrity of solid tissues.
Collapse
Affiliation(s)
- Caezar Al-Jassar
- School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | | | | | | |
Collapse
|
42
|
Chen YJ, Lee LY, Chao YK, Chang JT, Lu YC, Li HF, Chiu CC, Li YC, Li YL, Chiou JF, Cheng AJ. DSG3 facilitates cancer cell growth and invasion through the DSG3-plakoglobin-TCF/LEF-Myc/cyclin D1/MMP signaling pathway. PLoS One 2013; 8:e64088. [PMID: 23737966 PMCID: PMC3667790 DOI: 10.1371/journal.pone.0064088] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 04/10/2013] [Indexed: 02/04/2023] Open
Abstract
Desmoglein 3 (DSG3) is a component of the desmosome, which confers strong cell-cell adhesion. Previously, an oncogenic function of DSG3 has been found in head neck cancer (HNC). Here, we investigated how this molecule contributes to the malignant phenotype. Because DSG3 is associated with plakoglobin, we examined whether these phenotypic alterations were mediated through the plakoglobin molecule. Immunoprecipitation and immunofluorescence staining revealed that DSG3 silencing disrupted its interaction with plakoglobin and induced plakoglobin translocation from the cytoplasm to the nucleus. Knockdown of DSG3 significantly increased the interaction of plakoglobin with the transcriptional factor TCF and suppressed the TCF/LEF transcriptional activity. These effects further conferred to reduced expression of the TCF/LEF downstream target genes, including c-myc, cyclin D1, and MMP-7. Functional analyses showed that DSG3 silencing reduced cell growth and arrested cells at G0/G1 phase. Besides, cell migration and invasion abilities were also decreased. These cellular results were confirmed using tumor xenografts in mice, as DSG3 silencing led to the suppressed tumor growth, plakoglobin translocation and reduced expression of TCF/LEF target genes in tumors. Therefore, our study shows that the desmosomal protein DSG3 additionally functions to regulate malignant phenotypes via nuclear signaling. In conclusion, we found that DSG3 functions as an oncogene and facilitates cancer growth and invasion in HNC cells through the DSG3-plakoglobin-TCF/LEF pathway.
Collapse
Affiliation(s)
- Yin-Ju Chen
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan
- Translational Research Laboratory, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Medical Biotechnology, Chang Gung University, Taoyuan, Taiwan
| | - Li-Yu Lee
- Department of Pathology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yin-Ka Chao
- Department of Thoracic Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Joseph T. Chang
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ya-Ching Lu
- Department of Medical Biotechnology, Chang Gung University, Taoyuan, Taiwan
| | - Hsiao-Fang Li
- Department of Medical Biotechnology, Chang Gung University, Taoyuan, Taiwan
| | - Ching-Chi Chiu
- Department of Medical Biotechnology, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Chen Li
- Department of Medical Biotechnology, Chang Gung University, Taoyuan, Taiwan
| | - Yan-Liang Li
- Department of Medical Biotechnology, Chang Gung University, Taoyuan, Taiwan
| | - Jeng-Fong Chiou
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan
- Translational Research Laboratory, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Cancer Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Ann-Joy Cheng
- Department of Medical Biotechnology, Chang Gung University, Taoyuan, Taiwan
- * E-mail:
| |
Collapse
|
43
|
Griffin JR, Wriston CC, Peters MS, Lehman JS. Decreased expression of intercellular adhesion molecules in acantholytic squamous cell carcinoma compared with invasive well-differentiated squamous cell carcinoma of the skin. Am J Clin Pathol 2013; 139:442-7. [PMID: 23525614 DOI: 10.1309/ajcptn4bnjyiruwo] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Intercellular adhesion proteins are poorly characterized in acantholytic squamous cell carcinoma (ASCC), a more aggressive tumor than nonacantholytic invasive well-differentiated squamous cell carcinoma (SCC) of the skin. In this study we compared expression of Dsg3, E-cadherin, and syndecan-1 in ASCC and SCC. Immunohistochemical detection of Dsg3, E-cadherin, and syndecan-1 in 22 ASCCs and 22 SCCs was graded on a semiquantitative scale for intensity of staining (SI) and degree of circumferential staining (CS) about the cell membrane. Results were assessed by means of conditional logistic regression and χ(2) analysis. Dsg3 and E-cadherin expression (SI, CS) was significantly decreased (P < .05) in ASCC compared with SCC, whereas staining for syndecan-1 was similar in the 2 tumor types. Differences in expression of adhesion markers between ASCC and SCC may contribute to the development of acantholysis in ASCC and its more aggressive biologic behavior.
Collapse
Affiliation(s)
| | | | - Margot S. Peters
- Department of Dermatology, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Julia S. Lehman
- Department of Dermatology, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| |
Collapse
|
44
|
Wolf A, Rietscher K, Glaß M, Hüttelmaier S, Schutkowski M, Ihling C, Sinz A, Wingenfeld A, Mun A, Hatzfeld M. Insulin signaling via Akt2 switches plakophilin 1 function from stabilizing cell adhesion to promoting cell proliferation. J Cell Sci 2013; 126:1832-44. [PMID: 23444369 DOI: 10.1242/jcs.118992] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Downregulation of adherens junction proteins is a frequent event in carcinogenesis. How desmosomal proteins contribute to tumor formation by regulating the balance between adhesion and proliferation is not well understood. The desmosomal protein plakophilin 1 can increase intercellular adhesion by recruiting desmosomal proteins to the plasma membrane or stimulate proliferation by enhancing translation rates. Here, we show that these dual functions of plakophilin 1 are regulated by growth factor signaling. Insulin stimulation induced the phosphorylation of plakophilin 1, which correlated with reduced intercellular adhesion and an increased activity of plakophilin 1 in the stimulation of translation. Phosphorylation was mediated by Akt2 at four motifs within the plakophilin 1 N-terminal domain. A plakophilin 1 phospho-mimetic mutant revealed reduced intercellular adhesion and accumulated in the cytoplasm, where it increased translation and proliferation rates and conferred the capacity of anchorage-independent growth. The cytoplasmic accumulation was mediated by the stabilization of phosphorylated plakophilin 1, which displayed a considerably increased half-life, whereas non-phosphorylated plakophilin 1 was more rapidly degraded. Our data indicate that upon activation of growth factor signaling, plakophilin 1 switches from a desmosome-associated growth-inhibiting to a cytoplasmic proliferation-promoting function. This supports the view that the deregulation of plakophilin 1, as observed in several tumors, directly contributes to hyperproliferation and carcinogenesis in a context-dependent manner.
Collapse
Affiliation(s)
- Annika Wolf
- Institute of Molecular Medicine, Division of Pathobiochemistry, Martin-Luther-University Halle-Wittenberg, 06114 Halle, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kyrodimou M, Andreadis D, Drougou A, Amanatiadou EP, Angelis L, Barbatis C, Epivatianos A, Vizirianakis IS. Desmoglein-3/γ-catenin and E-cadherin/ß-catenin differential expression in oral leukoplakia and squamous cell carcinoma. Clin Oral Investig 2013; 18:199-210. [PMID: 23430339 DOI: 10.1007/s00784-013-0937-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 01/28/2013] [Indexed: 12/29/2022]
Abstract
OBJECTIVE The purpose of this study was to investigate gene/protein expression alterations of intercellular connections' components in oral leukoplakia (OLs) and squamous-cell carcinoma (OSCCs). MATERIALS AND METHODS Expression of desmogleins-2,3 (Dsg2/Dsg3), E-cadherin, and their cytoplasmic ligand, β/γ-catenins were quantitatively assessed in HSC-3 cells growing as monolayer cultures (ML)/multicellular aggregates (MCAs), using RT-PCR/Western blot, whereas their localization was detected by immunofluorescence. Furthermore, their expression was semi-quantitatively investigated in tissues from 25 OLs/25 OSCCs, using automated immunohistochemistry. RESULTS The steady-state levels of Dsg3 RNA transcripts increased as HSC-3 cells enter their exponential phase of growth, before a dramatic decrease to be observed as cells reached their plateau phase especially in MCAs. Upon the same period of time, Dsg2 levels have been increased. The expression of γ-catenin but not that of β-catenin was increased after 48 h in both MLs and MCAs. In clinical samples, Dsg3, Ε-cadherin, β/γ-catenin down-regulation was observed to be associated with the grade of OLs-dysplasia and OSCCs. Importantly, a membrane-to-cytoplasmic switch of expression and strong perinuclear aggregation of Dsg3/γ-catenin was seen in both HSC-3 cells and OLs/OSCCs. CONCLUSIONS The altered expression of Dsg3/γ-catenin and E-cadherin/β-catenin, in vitro and in ODs/OSCC imply their involvement in growth regulation and phenotype of dysplastic/malignant oral epithelial cells, contributing to the better understanding of epithelial dysplasia and OSCCs. CLINICAL RELEVANCE The observed alterations of their expression suggest a role of Dsg3 and γ-catenin (additionally to E-cadherin/β-catenin) as biomarkers of malignant transformation risk of oral dysplasia and the biological behavior (aggressiveness) of oral cancer, respectively.
Collapse
Affiliation(s)
- Marianthi Kyrodimou
- Department of Pharmaceutical Sciences, Laboratory of Pharmacology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Zanivan S, Meves A, Behrendt K, Schoof EM, Neilson LJ, Cox J, Tang HR, Kalna G, van Ree JH, van Deursen JM, Trempus CS, Machesky LM, Linding R, Wickström SA, Fässler R, Mann M. In vivo SILAC-based proteomics reveals phosphoproteome changes during mouse skin carcinogenesis. Cell Rep 2013; 3:552-66. [PMID: 23375375 DOI: 10.1016/j.celrep.2013.01.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 11/27/2012] [Accepted: 01/03/2013] [Indexed: 12/16/2022] Open
Abstract
Cancer progresses through distinct stages, and mouse models recapitulating traits of this progression are frequently used to explore genetic, morphological, and pharmacological aspects of tumor development. To complement genomic investigations of this process, we here quantify phosphoproteomic changes in skin cancer development using the SILAC mouse technology coupled to high-resolution mass spectrometry. We distill protein expression signatures from our data that distinguish between skin cancer stages. A distinct phosphoproteome of the two stages of cancer progression is identified that correlates with perturbed cell growth and implicates cell adhesion as a major driver of malignancy. Importantly, integrated analysis of phosphoproteomic data and prediction of kinase activity revealed PAK4-PKC/SRC network to be highly deregulated in SCC but not in papilloma. This detailed molecular picture, both at the proteome and phosphoproteome level, will prove useful for the study of mechanisms of tumor progression.
Collapse
Affiliation(s)
- Sara Zanivan
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ishige S, Kasamatsu A, Ogoshi K, Saito Y, Usukura K, Yokoe H, Kouzu Y, Koike H, Sakamoto Y, Ogawara K, Shiiba M, Tanzawa H, Uzawa K. Decreased expression of kallikrein-related peptidase 13: possible contribution to metastasis of human oral cancer. Mol Carcinog 2013; 53:557-65. [PMID: 23371469 DOI: 10.1002/mc.22007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 12/26/2012] [Accepted: 01/04/2013] [Indexed: 01/11/2023]
Abstract
The human kallikrein-related peptidase family is comprised of 15 serine protease genes on chromosome 19q13.4. Our previous microarray analyses showed that the gene kallikrein-related peptidase 13 (KLK13) was down-regulated in oral squamous cell carcinoma (OSCC) cell lines. We evaluated the expression status of KLK13 in primary OSCCs and performed functional molecular experiments in OSCC cell lines. In 102 primary tumors studied, KLK13 expression significantly (P < 0.05) decreased compared with matched normal counterparts. Interestingly, KLK13-negative cases correlated significantly (P < 0.05) with regional lymph node metastasis. In vitro, cells overexpressing KLK13 (oeKLK13) had decreased invasiveness and motility and up-regulation of adhesion molecules (E-cadherin, α-catenin, β-catenin, junction plakoglobin, plakophilin4, desmocollin2, desmoglein3, and desmoplakin) compared with control cells. A rescue experiment that transfected oeKLK13 cells with siRNA against KLK13 restored invasiveness and migration activities with down-regulated adhesion molecules. Based on our results, we concluded that KLK13 may play an important role in regulating cellular migration and invasiveness, making the loss of KLK13 a potential biomarker for early detection of lymph node metastasis in OSCCs.
Collapse
Affiliation(s)
- Shunsaku Ishige
- Department of Clinical Molecular Biology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Insights into the role of cell-cell junctions in physiology and disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 306:187-221. [PMID: 24016526 DOI: 10.1016/b978-0-12-407694-5.00005-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Contacting cells establish different classes of intricate structures at the cell-cell junctions. These structures are of increasing research interest as they regulate a broad variety of processes in development and disease. Further, in vitro studies are revealing that various cell-cell interaction proteins are involved not only in cell-cell processes but also in many additional aspects of physiology, such as migration and apoptosis. This chapter reviews the basic classification of cell-cell junctional structures and some of their representative proteins. Their roles in development and disease are briefly outlined, followed by a section on contemporary methods for probing cell-cell interactions and some recent developments. This chapter concludes with a few suggestions for potential research directions to further develop this promising area of study.
Collapse
|
49
|
Kyriakakis E, Maslova K, Philippova M, Pfaff D, Joshi MB, Buechner SA, Erne P, Resink TJ. T-Cadherin is an auxiliary negative regulator of EGFR pathway activity in cutaneous squamous cell carcinoma: impact on cell motility. J Invest Dermatol 2012; 132:2275-85. [PMID: 22592160 DOI: 10.1038/jid.2012.131] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Genetic and epigenetic studies in different cancers, including cutaneous carcinomas, have implicated T-cadherin (T-cad) as a tumor suppressor. Immunohistochemical and in vitro studies have suggested that T-cad loss promotes incipient invasiveness in cutaneous squamous cell carcinoma (SCC). Molecular mechanisms are unknown. This study found that the main consequence of T-cad silencing in SCC is facilitation of ligand-dependent EGFR activation, whereas T-cad overexpression impedes EGFR activation. Gain- and loss-of-function studies in A431 SCC cells demonstrate T-cad-controlled responsiveness to EGF with respect to pharmacological inhibition of EGFR and to diverse signaling and functional events of the EGFR activation cascade (EGFR phosphorylation, internalization, nuclear translocation, cell retraction/de-adhesion, motility, invasion, integrin β1, and Rho small GTPases such as RhoA, Rac1, and Cdc42 activation). Further, T-cad modulates the EGFR pathway activity by influencing membrane compartmentalization of EGFR; T-cad upregulation promotes retention of EGFR in lipid rafts, whereas T-cad silencing releases EGFR from this compartment, rendering EGFR more accessible to ligand stimulation. This study reveals a mechanism for fine-tuning of EGFR activity in SCC, whereby T-cad represents an auxiliary "negative" regulator of the EGFR pathway, which impacts invasion-associated behavioral responses of SCC to EGF. This action of T-cad in SCC may serve as a paradigm explaining other malignancies displaying concomitant T-cad loss and enhanced EGFR activity.
Collapse
Affiliation(s)
- Emmanouil Kyriakakis
- Laboratory for Signal Transduction, Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Beyer I, Cao H, Persson J, Song H, Richter M, Feng Q, Yumul R, van Rensburg R, Li Z, Berenson R, Carter D, Roffler S, Drescher C, Lieber A. Coadministration of epithelial junction opener JO-1 improves the efficacy and safety of chemotherapeutic drugs. Clin Cancer Res 2012; 18:3340-51. [PMID: 22535153 DOI: 10.1158/1078-0432.ccr-11-3213] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Epithelial junctions between tumor cells inhibit the penetration of anticancer drugs into tumors. We previously reported on recombinant adenovirus serotype 3-derived protein (JO-1), which triggers transient opening of intercellular junctions in epithelial tumors through binding to desmoglein 2 (DSG2), and enhances the antitumor effects of several therapeutic monoclonal antibodies. The goal of this study was to evaluate whether JO-1 cotherapy can also improve the efficacy of chemotherapeutic drugs. EXPERIMENTAL DESIGN The effect of intravenous application of JO-1 in combination with several chemotherapy drugs, including paclitaxel/Taxol, nanoparticle albumin-bound paclitaxel/Abraxane, liposomal doxorubicin/Doxil, and irinotecan/Camptosar, was tested in xenograft models for breast, colon, ovarian, gastric and lung cancer. Because JO-1 does not bind to mouse cells, for safety studies with JO-1, we also used human DSG2 (hDSG2) transgenic mice with tumors that overexpressed hDSG2. RESULTS JO-1 increased the efficacy of chemotherapeutic drugs, and in several models overcame drug resistance. JO-1 treatment also allowed for the reduction of drug doses required to achieve antitumor effects. Importantly, JO-1 coadmininstration protected normal tissues, including bone marrow and intestinal epithelium, against toxic effects that are normally associated with chemotherapeutic agents. Using the hDSG2-transgenic mouse model, we showed that JO-1 predominantly accumulates in tumors. Except for a mild, transient diarrhea, intravenous injection of JO-1 (2 mg/kg) had no critical side effects on other tissues or hematologic parameters in hDSG2-transgenic mice. CONCLUSIONS Our preliminary data suggest that JO-1 cotherapy has the potential to improve the therapeutic outcome of cancer chemotherapy.
Collapse
Affiliation(s)
- Ines Beyer
- Division of Medical Genetics, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|