1
|
Sun H, Liang Z, Zang Y, Liu S, Liu H, Li M. Transcriptome Analysis Reveals Possible Antitumor Mechanism of Intracellular Polysaccharide From Phaeodactylum tricornutum on Cervical Cancer HeLa Cells. Chem Biodivers 2025:e202402779. [PMID: 40251892 DOI: 10.1002/cbdv.202402779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
Seaweed polysaccharide, a naturally occurring, non-toxic antitumor substance, has emerged as a significant focus of research. In this study, intracellular polysaccharides from Phaeodactylum tricornutum (PRP) were isolated and purified to investigate their antitumor effects and underlying mechanisms. The inhibitory effects of various purified polysaccharide fractions on cervical cancer cells were evaluated, and their antitumor mechanisms were elucidated through transcriptome analysis. The results demonstrated that all four purified polysaccharide fractions from P. tricornutum inhibited HeLa cell proliferation, reduced cell viability, and altered cell morphology. According to the cell counting kit-8 (CCK-8) assay, PRP4 exhibited the most potent inhibitory effect among the four fractions. Transcriptome analysis revealed 806 differentially expressed genes (DEGs) in the PRP4-treated group compared to the control, comprising 570 up-regulated and 236 down-regulated genes. Gene function enrichment analysis indicated that DEGs were significantly enriched in apoptosis- and tumor-related biological processes, implicating multiple cancer- and apoptosis-associated signaling pathways. A protein-protein interaction (PPI) network identified 10 DEGs as hub genes, namely TLR4, interleukin-1β (IL1B), heme oxygenase-1 (HMOX1), EDN1, PTGS2, MMP9, CXCL8, TGFB1, connective tissue growth factor (CTGF), and SERPINE1. These findings suggest that PRP4 holds promise as a therapeutic agent for cancer treatment.
Collapse
Affiliation(s)
- Han Sun
- Guangxi Key Laboratory of Polysaccharide Materials and Modification, School of Marine and Biotechnology, Guangxi Minzu University, Nanning, China
- Huangshan Vocational and Technical College, Huangshan City, China
| | - Zhongwen Liang
- Guangxi Key Laboratory of Polysaccharide Materials and Modification, School of Marine and Biotechnology, Guangxi Minzu University, Nanning, China
| | - Ying Zang
- Guangxi Key Laboratory of Polysaccharide Materials and Modification, School of Marine and Biotechnology, Guangxi Minzu University, Nanning, China
| | - Song Liu
- Guangxi Key Laboratory of Polysaccharide Materials and Modification, School of Marine and Biotechnology, Guangxi Minzu University, Nanning, China
| | - Hongquan Liu
- Guangxi Key Laboratory of Polysaccharide Materials and Modification, School of Marine and Biotechnology, Guangxi Minzu University, Nanning, China
| | - Mei Li
- Guangxi Key Laboratory of Polysaccharide Materials and Modification, School of Marine and Biotechnology, Guangxi Minzu University, Nanning, China
| |
Collapse
|
2
|
Johnson B, Iuliano M, Lam TT, Biederer T, De Camilli PV. A complex of the lipid transport ER proteins TMEM24 and C2CD2 with band 4.1 at cell-cell contacts. J Cell Biol 2024; 223:e202311137. [PMID: 39158698 PMCID: PMC11334333 DOI: 10.1083/jcb.202311137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/23/2024] [Accepted: 08/07/2024] [Indexed: 08/20/2024] Open
Abstract
Junctions between the ER and plasma membrane (PM) are implicated in calcium homeostasis, non-vesicular lipid transfer, and other cellular functions. Two ER proteins that function both as tethers to the PM via a polybasic C-terminus motif and as phospholipid transporters are brain-enriched TMEM24 (C2CD2L) and its paralog C2CD2. We report that both proteins also form a complex with band 4.1 family members, which in turn bind PM proteins including cell adhesion molecules such as SynCAM 1. This complex enriches TMEM24 and C2CD2 containing ER/PM junctions at sites of cell contacts. Dynamic properties of TMEM24-dependent ER/PM junctions are impacted when band 4.1 is part of the junction, as TMEM24 at cell-adjacent ER/PM junctions is not shed from the PM by calcium rise, unlike TMEM24 at non-cell adjacent junctions. Lipid transport between the ER and the PM by TMEM24 and C2CD2 at sites where cells, including neurons, contact other cells may participate in adaptive responses to cell contact-dependent signaling.
Collapse
Affiliation(s)
- Ben Johnson
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Maria Iuliano
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - TuKiet T Lam
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT, USA
- Department of Keck MS and Proteomics Resource, Yale University School of Medicine, New Haven, CT, USA
| | - Thomas Biederer
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
| | - Pietro V De Camilli
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
3
|
Koster AK, Yarishkin O, Dubin AE, Kefauver JM, Pak RA, Cravatt BF, Patapoutian A. Chemical mapping of the surface interactome of PIEZO1 identifies CADM1 as a modulator of channel inactivation. Proc Natl Acad Sci U S A 2024; 121:e2415934121. [PMID: 39356664 PMCID: PMC11474052 DOI: 10.1073/pnas.2415934121] [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: 08/07/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
The propeller-shaped blades of the PIEZO1 and PIEZO2 ion channels partition into the plasma membrane and respond to indentation or stretching of the lipid bilayer, thus converting mechanical forces into signals that can be interpreted by cells, in the form of calcium flux and changes in membrane potential. While PIEZO channels participate in diverse physiological processes, from sensing the shear stress of blood flow in the vasculature to detecting touch through mechanoreceptors in the skin, the molecular details that enable these mechanosensors to tune their responses over a vast dynamic range of forces remain largely uncharacterized. To survey the molecular landscape surrounding PIEZO channels at the cell surface, we employed a mass spectrometry-based proteomic approach to capture and identify extracellularly exposed proteins in the vicinity of PIEZO1. This PIEZO1-proximal interactome was enriched in surface proteins localized to cell junctions and signaling hubs within the plasma membrane. Functional screening of these interaction candidates by calcium imaging and electrophysiology in an overexpression system identified the adhesion molecule CADM1/SynCAM that slows the inactivation kinetics of PIEZO1 with little effect on PIEZO2. Conversely, we found that CADM1 knockdown accelerates inactivation of endogenous PIEZO1 in Neuro-2a cells. Systematic deletion of CADM1 domains indicates that the transmembrane region is critical for the observed effects on PIEZO1, suggesting that modulation of inactivation is mediated by interactions in or near the lipid bilayer.
Collapse
Affiliation(s)
- Anna K. Koster
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
- Department of Chemistry, Scripps Research, La Jolla, CA92037
| | - Oleg Yarishkin
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| | - Adrienne E. Dubin
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| | - Jennifer M. Kefauver
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| | - Ryan A. Pak
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| | | | - Ardem Patapoutian
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| |
Collapse
|
4
|
Hagiyama M, Yoneshige A, Wada A, Kimura R, Ito S, Inoue T, Takeuchi F, Ito A. Efficient intracellular drug delivery by co-administration of two antibodies against cell adhesion molecule 1. J Control Release 2024; 371:603-618. [PMID: 38782061 DOI: 10.1016/j.jconrel.2024.05.035] [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: 01/10/2024] [Revised: 05/02/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024]
Abstract
Cell adhesion molecule 1 (CADM1), a single-pass transmembrane protein, is involved in oncogenesis. We previously demonstrated the therapeutic efficacy of anti-CADM1 ectodomain monoclonal antibodies against mesothelioma; however, the underlying mechanism is unclear. In the present study, we explored the molecular behavior of anti-CADM1 antibodies in CADM1-expressing tumor cells. Sequencing analyses revealed that the anti-CADM1 chicken monoclonal antibodies 3E1 and 9D2 are IgY and IgM isotype antibodies, respectively. Co-administration of 3E1 and 9D2 altered the subcellular distribution of CADM1 from the detergent-soluble fraction to the detergent-resistant fraction in tumor cells. Using recombinant chicken-mouse chimeric antibodies that had been isotype-switched from IgG to IgM, we demonstrated that the combination of the variable region of 3E1 and the constant region of IgM was required for CADM1 relocation. Cytochemical studies showed that 3E1 colocalized with late endosomes/lysosomes after co-administration with 9D2, suggesting that the CADM1-antibody complex is internalized from the cell surface to intracellular compartments by lipid-raft mediated endocytosis. Finally, 3E1 was conjugated with the antimitotic agent monomethyl auristatin E (MMAE) via a cathepsin-cleavable linker. Co-administration of 3E1-monomethyl auristatin E and 9D2 suppressed the growth of multiple types of tumor cells, and this anti-tumor activity was confirmed in a syngeneic mouse model of melanoma. 3E1 and 9D2 are promising drug delivery vehicles for CADM1-expressing tumor cells.
Collapse
Affiliation(s)
- Man Hagiyama
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Azusa Yoneshige
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan.
| | - Akihiro Wada
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Ryuichiro Kimura
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Shinji Ito
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takao Inoue
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Fuka Takeuchi
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Akihiko Ito
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan.
| |
Collapse
|
5
|
Johnson B, Iuliano M, Lam T, Biederer T, De Camilli P. A complex of the lipid transport ER proteins TMEM24 and C2CD2 with band 4.1 at cell-cell contacts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.06.570396. [PMID: 38106008 PMCID: PMC10723409 DOI: 10.1101/2023.12.06.570396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Junctions between the ER and the plasma membrane (ER/PM junctions) are implicated in calcium homeostasis, non-vesicular lipid transfer and other cellular functions. Two ER proteins that function both as membrane tethers to the PM via a polybasic motif in their C-terminus and as phospholipid transporters are brain-enriched TMEM24 (C2CD2L) and its paralog C2CD2. Based on an unbiased proximity ligation analysis, we found that both proteins can also form a complex with band 4.1 family members, which in turn can bind a variety of plasma membrane proteins including cell adhesion molecules such as SynCAM 1. This complex results in the enrichment of TMEM24 and C2CD2 containing ER/PM junctions at sites of cell contacts. Dynamic properties of TMEM24-dependent ER/PM contacts are impacted when in complex as TMEM24 present at cell adjacent junctions is not shed by calcium rise, unlike TMEM24 at non-cell adjacent junctions. These findings suggest that cell-contact interactions control ER/PM junctions via TMEM24 complexes involving band 4.1 proteins.
Collapse
Affiliation(s)
- Ben Johnson
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Maria Iuliano
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111
| | - TuKiet Lam
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Keck MS and Proteomics Resource, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Thomas Biederer
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Pietro De Camilli
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| |
Collapse
|
6
|
Liu Z, Xu Y, Jin S, Liu X, Wang B. Construction of a Prognostic Model Based on Methylation-Related Genes in Patients with Colon Adenocarcinoma. Cancer Manag Res 2023; 15:1097-1110. [PMID: 37818334 PMCID: PMC10561619 DOI: 10.2147/cmar.s417897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 09/22/2023] [Indexed: 10/12/2023] Open
Abstract
Purpose Colon adenocarcinoma (COAD) is the second leading cause of death in the world, and the new incidence rate ranks third among all cancers. Abnormal DNA methylation is related to the occurrence and development of tumors. In this study, we aimed to identify genes associated with abnormal methylation in COAD. Methods COAD transcriptome data, methylation data and clinical information were downloaded from the TCGA database and GEO database. The differentially expressed genes (DEGs) and methylated genes (DMGs) were analyzed and identified in COAD. PCA analysis was applied to divide COAD into subtypes, and the survival and immune cell infiltration of each subtype were evaluated. Cox and LASSO analyses were performed to construct COAD risk model. GSEA was used to evaluate the enrichment pathways. The Kaplan-Meier was used to analyze the difference in survival. ROC curve was plotted to evaluate the accuracy of the model, and GSE17536 was used to verify the accuracy of the risk model. The risk model is combined with the clinicopathological characteristics of COAD patients to perform multivariate Cox regression analysis to obtain independent risk factors and draw nomograms. Results In total, 4564 DEGs and 1093 DMGs were screened, among which 298 were found to be overlapping genes. For 220 of these overlapping genes, the methylation was significantly negatively correlated to expression levels. An optimal signature from 4 methylated biomarkers was identified to construct the prognostic model. Conclusion Our study identified 4 methylated biomarkers in the COAD. Then, we constructed the risk model to provide a theoretical basis and reference value for the research and treatment of COAD.
Collapse
Affiliation(s)
- ZhenDong Liu
- Department of General Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| | - YuYang Xu
- Department of General Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| | - Shan Jin
- Department of Anesthesiology, Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| | - Xin Liu
- Department of General Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| | - BaoChun Wang
- Department of General Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| |
Collapse
|
7
|
Hagiyama M, Mimae T, Wada A, Takeuchi F, Yoneshige A, Inoue T, Kotoku N, Hamada H, Sekido Y, Okada M, Ito A. Possible Therapeutic Utility of anti-Cell Adhesion Molecule 1 Antibodies for Malignant Pleural Mesothelioma. Front Cell Dev Biol 2022; 10:945007. [PMID: 35903548 PMCID: PMC9315061 DOI: 10.3389/fcell.2022.945007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/23/2022] [Indexed: 11/15/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a highly aggressive malignant tumor, and the effective therapeutic drugs are limited. Thus, the establishment of novel therapeutic method is desired. Considerable proportion of MPMs are shown to express cell adhesion molecule 1 (CADM1), and to use CADM1 to bind to and proliferate on the pleural mesothelial surface, suggesting that CADM1 is a possible therapeutic target. Here, anti-CADM1 ectodomain chicken monoclonal antibodies, 3E1 and 9D2, were examined for their possible therapeutic utility. The full-length form of CADM1 was expressed in eight out of twelve human MPM cell lines. MPM cell lines were cultured on a confluent monolayer of mesothelial MeT-5A cells in the presence of 9D2, the neutralizing antibody. 9D2 suppressed the cell growth of CADM1-positive MPM cells with the loss and aggregation of CADM1 molecules on the MPM cell membrane, but not of CADM1-negative MPM cells. Co-addition of 3E1, lacking the neutralizing action, enhanced the growth-suppressive effect of 9D2. The two antibodies were tested as drug delivery vectors. 3E1 was converted into a humanized antibody (h3E1) and conjugated with monomethyl auristatin E (MMAE), a tubulin polymerization inhibitor. When the resulting h3E1–MMAE antibody-drug conjugate (ADC) was added to the standard cultures of CADM1-positive MPM cells, it suppressed the cell growth in a dose-dependent manner. Co-addition of 9D2 enhanced the growth-suppressive effect of h3E1–MMAE ADC. Anti-CADM1 ectodomain antibodies were suggested to serve as both antibody drugs and drug vectors in the treatment of MPM.
Collapse
Affiliation(s)
- Man Hagiyama
- Department of Pathology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Takahiro Mimae
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Akihiro Wada
- Department of Pathology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Fuka Takeuchi
- Division of Molecular Pathology, Graduate School of Medical Science, Kindai University, Osaka, Japan
| | - Azusa Yoneshige
- Department of Pathology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Takao Inoue
- Department of Pathology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Naoyuki Kotoku
- College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan
| | - Hironobu Hamada
- Department of Physical Analysis and Therapeutic Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yoshitaka Sekido
- Division of Cancer Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Morihito Okada
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Akihiko Ito
- Department of Pathology, Kindai University Faculty of Medicine, Osaka, Japan
- Division of Molecular Pathology, Graduate School of Medical Science, Kindai University, Osaka, Japan
- *Correspondence: Akihiko Ito,
| |
Collapse
|
8
|
miR-486 Promotes the Invasion and Cell Cycle Progression of Ovarian Cancer Cells by Targeting CADM1. ACTA ACUST UNITED AC 2021; 2021:7407086. [PMID: 34395181 PMCID: PMC8360751 DOI: 10.1155/2021/7407086] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/13/2021] [Indexed: 01/05/2023]
Abstract
Objective To explore the role and possible underlying mechanism of miR-486 in ovarian cancer (OC) cells. Methods The expression of miR-486 and CADM1 was detected by qRT-PCR in OC tissues and adjacent nontumor tissues and OC cell lines. The dual-luciferase reporter gene system was used to determine the targeting relationship between miR-486 and CADM1. CCK-8, colony formation assay, Transwell, and flow cytometry were performed to detect cell proliferation, cell invasion, cell cycle progression, and the apoptotic cell death, respectively. Western blot was carried out to detect the expression of CADM1 protein and the proteins associated with cell cycle progression. Results miR-486 was significantly upregulated in OC tissues and cells, while CADM1 expression was significantly downregulated. Dual-luciferase reporter assays further confirmed that CADM1 was a target gene of miR-486. Interference with miR-486 could inhibit the proliferation and invasion and promoted the apoptosis of SKOV3 cells. Knocking down both miR-486 and CADM1 significantly increased the SKOV3 cell proliferation, invasion, and the number of cells transitioning from the G0/G1 phase into the S phase of cell cycle and reduced the cellular apoptosis. Western blot analysis revealed that the expression of cell cycle progression-related proteins (CyclinD1, CyclinE, and CDK6) was significantly reduced, and the p21 expression was increased when interfering with both miR-486 and CADM1 expression. Conclusion Our results suggested that miR-486 could act as a tumor promoter by targeting CADM1 and be a potential therapeutic target for the treatment of OC.
Collapse
|
9
|
Li H, Gao J, Zhang S. Functional and Clinical Characteristics of Cell Adhesion Molecule CADM1 in Cancer. Front Cell Dev Biol 2021; 9:714298. [PMID: 34395444 PMCID: PMC8361327 DOI: 10.3389/fcell.2021.714298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/02/2021] [Indexed: 12/21/2022] Open
Abstract
The cell adhesion molecule CADM1, which participates in cell adhesion and signal transduction, has a regulatory effect on the development of tumors. CADM1 is often involved in malignant tumors of multiple organ systems, such as the respiratory and digestive systems. Upregulated CADM1 promotes tumor cell apoptosis and inhibits malignant proliferation. Along with cell cycle-related proteins, it participates in regulating signaling pathways, such as EMT, STAT3, and AKT, and plays an important role in inhibiting invasion and migration. Considering clinical characteristics, low CADM1 expression is associated with aggressive tumors and poor prognosis. In addition, some long non-coding RNAs (lncRNAs) or miRNAs directly or indirectly act on CADM1 to regulate tumor growth and motility. Interestingly, CADM1 function differs in adult T-cell leukemia/lymphoma (ATLL), and NF-κB is thought to be involved in this process. Taken together, CADM1 could be a potential biomarker for early diagnosis and a target for cancer treatment in future clinical practices.
Collapse
Affiliation(s)
- Hongxu Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ, Transplantation at Henan Universities, Zhengzhou, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Jie Gao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ, Transplantation at Henan Universities, Zhengzhou, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Shuijun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ, Transplantation at Henan Universities, Zhengzhou, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| |
Collapse
|
10
|
Kimura R, Otani T, Shiraishi N, Hagiyama M, Yoneshige A, Wada A, Kajiyama H, Takeuchi F, Mizuguchi N, Morishita K, Ito A. Expression of cell adhesion molecule 1 in human and murine endometrial glandular cells and its increase during the proliferative phase by estrogen and cell density. Life Sci 2021; 283:119854. [PMID: 34332980 DOI: 10.1016/j.lfs.2021.119854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/14/2021] [Accepted: 07/22/2021] [Indexed: 10/20/2022]
Abstract
AIMS Cell adhesion molecule 1 (CADM1) mediates interepithelial adhesion and is upregulated in crowded epithelial monolayers. This study aimed to examine CADM1 expression in the human endometrium of proliferative and secretory phases, and its transcriptional regulation in terms of estrogen stimuli and higher cellularity. MAIN METHODS CADM1 immunohistochemistry was conducted on endometrial tissues from women in their 40s and adult mice subcutaneously injected with estradiol following ovariectomy. Dual-luciferase reporter assays were conducted using human endometrial HEC-50B and HEC-1B cells and reporter plasmids harboring the human CADM1 3.4-kb promoter and its deleted and mutated forms. Cells were transfected with estrogen receptor α cDNA and reporter plasmids, and treated with estradiol before luciferase activity measurement. KEY FINDINGS Immunohistochemistry revealed that CADM1 was clearly expressed on the lateral membranes of the simple columnar glandular cells in the proliferative phase, but not in the secretory phase, from both women and the mouse model. The glandular cell density increased two-fold in the proliferative phase. Reporter assays identified three Sp1-binding sites as estradiol-responsive elements in the proximal region (from -223 to -84) of the transcription start site (+1) in HEC-50B cells. When the cell culture was started at eight-fold higher cell density, the CADM1 3.4-kb promoter was transactivated at a two-fold higher level in HEC-50B cells. This cell density effect was not detected for the CADM1 2.3-kb or 1.6-kb promoter. SIGNIFICANCE Two (proximal and distal) promoter regions are suggested to function additively to transactivate CADM1 in endometrial glandular cells that crowd in the proliferative phase.
Collapse
Affiliation(s)
- Ryuichiro Kimura
- Department of Pathology, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka 589-8511, Japan
| | - Tomoyuki Otani
- Department of Pathology, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka 589-8511, Japan
| | - Naoki Shiraishi
- Genome Medical Center, Kindai University Hospital, Osaka, Japan
| | - Man Hagiyama
- Department of Pathology, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka 589-8511, Japan
| | - Azusa Yoneshige
- Department of Pathology, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka 589-8511, Japan
| | - Akihiro Wada
- Department of Pathology, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka 589-8511, Japan
| | - Hiroshi Kajiyama
- Department of Pathology, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka 589-8511, Japan
| | - Fuka Takeuchi
- Department of Pathology, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka 589-8511, Japan
| | | | - Kazuhiro Morishita
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Akihiko Ito
- Department of Pathology, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka 589-8511, Japan.
| |
Collapse
|
11
|
Funaki T, Ito T, Tanei ZI, Goto A, Niki T, Matsubara D, Murakami Y. CADM1 promotes malignant features of small-cell lung cancer by recruiting 4.1R to the plasma membrane. Biochem Biophys Res Commun 2021; 534:172-178. [PMID: 33298314 DOI: 10.1016/j.bbrc.2020.11.121] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023]
Abstract
Cell adhesion molecule 1 (CADM1), which mediates intercellular adhesion between epithelial cells, is shown to be highly expressed in small-cell lung cancer (SCLC) and to enhance tumorigenicity of SCLC cells in nude mice. Here, we investigated the molecular mechanism underlying the oncogenic role of CADM1 in SCLC. CADM1 promoted colony formation of SCLC cells in soft agar. Analysis of deletion and point mutants of the conserved protein-binding motifs in CADM1 revealed that the 4.1 protein-binding motif in the cytoplasmic domain is responsible for the promotion of colony formation. Among the actin-binding 4.1 proteins, 4.1R was the only protein whose localization to the plasma membrane is dependent on CADM1 expression in SCLC cells. Knockdown of 4.1R suppressed the colony formation enhanced by CADM1, suggesting that 4.1R is required for the oncogenic role of CADM1 in SCLC. In primary SCLC, CADM1 expression was correlated with membranous localization of 4.1R, as was observed in a SCLC cell line. Moreover, membranous co-localization of CADM1 and 4.1R was associated with more advanced tumor stage. These results suggest that the formation of CADM1-4.1R complex would promote malignant features of SCLC.
Collapse
Affiliation(s)
- Toko Funaki
- Division of Molecular Pathology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takeshi Ito
- Division of Molecular Pathology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Zen-Ichi Tanei
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Akiteru Goto
- Department of Cellular and Organ Pathology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Toshiro Niki
- Department of Integrative Pathology, Jichi Medical University, Shimotsuke, Japan
| | - Daisuke Matsubara
- Division of Molecular Pathology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Integrative Pathology, Jichi Medical University, Shimotsuke, Japan
| | - Yoshinori Murakami
- Division of Molecular Pathology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
12
|
Sanders LM, Cheney A, Seninge L, van den Bout A, Chen M, Beale HC, Kephart ET, Pfeil J, Learned K, Lyle AG, Bjork I, Haussler D, Salama SR, Vaske OM. Identification of a differentiation stall in epithelial mesenchymal transition in histone H3-mutant diffuse midline glioma. Gigascience 2020; 9:giaa136. [PMID: 33319914 PMCID: PMC7736793 DOI: 10.1093/gigascience/giaa136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/17/2020] [Accepted: 11/05/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Diffuse midline gliomas with histone H3 K27M (H3K27M) mutations occur in early childhood and are marked by an invasive phenotype and global decrease in H3K27me3, an epigenetic mark that regulates differentiation and development. H3K27M mutation timing and effect on early embryonic brain development are not fully characterized. RESULTS We analyzed multiple publicly available RNA sequencing datasets to identify differentially expressed genes between H3K27M and non-K27M pediatric gliomas. We found that genes involved in the epithelial-mesenchymal transition (EMT) were significantly overrepresented among differentially expressed genes. Overall, the expression of pre-EMT genes was increased in the H3K27M tumors as compared to non-K27M tumors, while the expression of post-EMT genes was decreased. We hypothesized that H3K27M may contribute to gliomagenesis by stalling an EMT required for early brain development, and evaluated this hypothesis by using another publicly available dataset of single-cell and bulk RNA sequencing data from developing cerebral organoids. This analysis revealed similarities between H3K27M tumors and pre-EMT normal brain cells. Finally, a previously published single-cell RNA sequencing dataset of H3K27M and non-K27M gliomas revealed subgroups of cells at different stages of EMT. In particular, H3.1K27M tumors resemble a later EMT stage compared to H3.3K27M tumors. CONCLUSIONS Our data analyses indicate that this mutation may be associated with a differentiation stall evident from the failure to proceed through the EMT-like developmental processes, and that H3K27M cells preferentially exist in a pre-EMT cell phenotype. This study demonstrates how novel biological insights could be derived from combined analysis of several previously published datasets, highlighting the importance of making genomic data available to the community in a timely manner.
Collapse
Affiliation(s)
- Lauren M Sanders
- Department of Biomolecular Engineering, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
- University of California Santa Cruz Genomics Institute, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Allison Cheney
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Lucas Seninge
- Department of Biomolecular Engineering, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
- University of California Santa Cruz Genomics Institute, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Anouk van den Bout
- University of California Santa Cruz Genomics Institute, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Marissa Chen
- University of California Santa Cruz Genomics Institute, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Holly C Beale
- University of California Santa Cruz Genomics Institute, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Ellen Towle Kephart
- University of California Santa Cruz Genomics Institute, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Jacob Pfeil
- Department of Biomolecular Engineering, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
- University of California Santa Cruz Genomics Institute, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Katrina Learned
- University of California Santa Cruz Genomics Institute, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - A Geoffrey Lyle
- University of California Santa Cruz Genomics Institute, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Isabel Bjork
- University of California Santa Cruz Genomics Institute, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - David Haussler
- Department of Biomolecular Engineering, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
- University of California Santa Cruz Genomics Institute, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
- Howard Hughes Medical Institute, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Sofie R Salama
- Department of Biomolecular Engineering, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
- University of California Santa Cruz Genomics Institute, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
- Howard Hughes Medical Institute, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Olena M Vaske
- University of California Santa Cruz Genomics Institute, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| |
Collapse
|
13
|
Chytła A, Gajdzik-Nowak W, Olszewska P, Biernatowska A, Sikorski AF, Czogalla A. Not Just Another Scaffolding Protein Family: The Multifaceted MPPs. Molecules 2020; 25:molecules25214954. [PMID: 33114686 PMCID: PMC7662862 DOI: 10.3390/molecules25214954] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 01/03/2023] Open
Abstract
Membrane palmitoylated proteins (MPPs) are a subfamily of a larger group of multidomain proteins, namely, membrane-associated guanylate kinases (MAGUKs). The ubiquitous expression and multidomain structure of MPPs provide the ability to form diverse protein complexes at the cell membranes, which are involved in a wide range of cellular processes, including establishing the proper cell structure, polarity and cell adhesion. The formation of MPP-dependent complexes in various cell types seems to be based on similar principles, but involves members of different protein groups, such as 4.1-ezrin-radixin-moesin (FERM) domain-containing proteins, polarity proteins or other MAGUKs, showing their multifaceted nature. In this review, we discuss the function of the MPP family in the formation of multiple protein complexes. Notably, we depict their significant role for cell physiology, as the loss of interactions between proteins involved in the complex has a variety of negative consequences. Moreover, based on recent studies concerning the mechanism of membrane raft formation, we shed new light on a possible role played by MPPs in lateral membrane organization.
Collapse
Affiliation(s)
- Agnieszka Chytła
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.C.); (W.G.-N.); (P.O.); (A.B.)
| | - Weronika Gajdzik-Nowak
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.C.); (W.G.-N.); (P.O.); (A.B.)
| | - Paulina Olszewska
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.C.); (W.G.-N.); (P.O.); (A.B.)
| | - Agnieszka Biernatowska
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.C.); (W.G.-N.); (P.O.); (A.B.)
| | - Aleksander F. Sikorski
- Research and Development Center, Regional Specialist Hospital, Kamieńskiego 73a, 51-154 Wroclaw, Poland;
| | - Aleksander Czogalla
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.C.); (W.G.-N.); (P.O.); (A.B.)
- Correspondence: ; Tel.: +48-71375-6356
| |
Collapse
|
14
|
Tsuboi Y, Oyama M, Kozuka-Hata H, Ito A, Matsubara D, Murakami Y. CADM1 suppresses c-Src activation by binding with Cbp on membrane lipid rafts and intervenes colon carcinogenesis. Biochem Biophys Res Commun 2020; 529:854-860. [PMID: 32616310 DOI: 10.1016/j.bbrc.2020.05.103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 05/14/2020] [Indexed: 12/16/2022]
Abstract
Cell adhesion molecules act as tumor suppressors primarily by cell attachment activity, but additional mechanisms modifying signal transduction are suggested in some cases. Cell adhesion molecule 1 (CADM1), a membrane-spanning immunoglobulin superfamily, mediates intercellular adhesion by trans-homophilic interaction and acts as a tumor suppressor. Here, we investigated CADM1-associated proteins comprehensively using proteomic analysis of immune-precipitates of CADM1 by mass spectrometry and identified a transmembrane adaptor protein, Csk-binding protein (Cbp), known to suppress Src-mediated transformation, as a binding partner of CADM1. CADM1 localizes to detergent-resistant membrane fractions and co-immunoprecipitated with Cbp and c-Src. Suppression of CADM1 expression using siRNA reduces the amount of co-immunoprecipitated c-Src with Cbp and activates c-Src in colon cancer cells expressing both CADM1 and Cbp. On the other hand, co-replacement of CADM1 and Cbp in colon cancer cells lacking CADM1 and Cbp expression suppresses c-Src activation, wound healing and tumorigenicity in nude mice. Furthermore, expression of Cbp and CADM1 was lost in 55% and 83% of human colon cancer, respectively, preferentially in tumors with larger size and/or lymph node metastasis. CADM1 would act as a colon tumor suppressor by intervening oncogenic c-Src signaling through binding with Cbp besides its authentic cell adhesion activity.
Collapse
Affiliation(s)
- Yumi Tsuboi
- Division of Molecular Pathology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masaaki Oyama
- Medical Proteomics Laboratory, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiroko Kozuka-Hata
- Medical Proteomics Laboratory, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Akihiko Ito
- Department of Pathology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Daisuke Matsubara
- Division of Integrative Pathology, Jichii Medical University, Shimotsuke, Japan
| | - Yoshinori Murakami
- Division of Molecular Pathology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
15
|
Hagiyama M, Kimura R, Yoneshige A, Inoue T, Otani T, Ito A. Cell Adhesion Molecule 1 Contributes to Cell Survival in Crowded Epithelial Monolayers. Int J Mol Sci 2020; 21:ijms21114123. [PMID: 32527032 PMCID: PMC7312920 DOI: 10.3390/ijms21114123] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/02/2020] [Accepted: 06/06/2020] [Indexed: 12/14/2022] Open
Abstract
When epithelial cells in vivo are stimulated to proliferate, they crowd and often grow in height. These processes are likely to implicate dynamic interactions among lateral membranous proteins, such as cell adhesion molecule 1 (CADM1), an immunoglobulin superfamily member. Pulmonary epithelial cell lines that express CADM1, named NCI-H441 and RLE-6TN, were grown to become overconfluent in the polarized 2D culture system, and were examined for the expression of CADM1. Western analyses showed that the CADM1 expression levels increased gradually up to 3 times in a cell density-dependent manner. Confocal microscopic observations revealed dense immunostaining for CADM1 on the lateral membrane. In the overconfluent monolayers, CADM1 knockdown was achieved by two methods using CADM1-targeting siRNA and an anti-CADM1 neutralizing antibody. Antibody treatment experiments were also done on 6 other epithelial cell lines expressing CADM1. The CADM1 expression levels were reduced roughly by half, in association with cell height decrease by half in 3 lines. TUNEL assays revealed that the CADM1 knockdown increased the proportion of TUNEL-positive apoptotic cells approximately 10 folds. Increased expression of CADM1 appeared to contribute to cell survival in crowded epithelial monolayers.
Collapse
|
16
|
Yuan X, Piao L, Wang L, Han X, Zhuang M, Liu Z. Pivotal roles of protein 4.1B/DAL‑1, a FERM‑domain containing protein, in tumor progression (Review). Int J Oncol 2019; 55:979-987. [PMID: 31545421 DOI: 10.3892/ijo.2019.4877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/04/2019] [Indexed: 11/06/2022] Open
Abstract
Protein 4.1B/DAL‑1, encoded by erythrocyte membrane protein band 4.1‑like 3 (EPB41L3), belongs to the protein 4.1 superfamily, a group of proteins that share a conserved four.one‑ezrin‑radixin‑moesin (FERM) domain. Protein 4.1B/DAL‑1 serves a crucial role in cytoskeletal organization and a number of processes through multiple interactions with membrane proteins via its FERM, spectrin‑actin‑binding and C‑terminal domains. A number of studies have indicated that a loss of EPB41L3 expression is commonly observed in lung cancer, breast cancer, esophageal squamous cell carcinoma and meningiomas. DNA methylation and a loss of heterozygosity have been reported to contribute to the downregulation of EPB41L3. To date, the biological functions of protein 4.1B/DAL‑1 in carcinogenesis remain unknown. The present review summarizes the current understanding of the role of protein 4.1B/DAL‑1 in cancer and highlights its potential as a cancer diagnostic and prognostic biomarker in cancer therapeutics.
Collapse
Affiliation(s)
- Xiaofeng Yuan
- Department of Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Lianhua Piao
- Institute of Bioinformatics and Medical Engineering, Jiangsu University of Technology, Changzhou, Jiangsu 213001, P.R. China
| | - Luhui Wang
- Department of Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Xu Han
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Ming Zhuang
- Department of Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Zhiwei Liu
- Department of Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| |
Collapse
|
17
|
Xue F, An C, Chen L, Liu G, Ren F, Guo X, Sun H, Mei L, Sun X, Li J, Tang Y, An X, Zheng P. 4.1B suppresses cancer cell proliferation by binding to EGFR P13 region of intracellular juxtamembrane segment. Cell Commun Signal 2019; 17:115. [PMID: 31492173 PMCID: PMC6731589 DOI: 10.1186/s12964-019-0431-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/29/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Gastric cancer (GC) has high incidence and mortality worldwide. However, the underlying mechanisms that regulate gastric carcinogenesis are largely undefined. 4.1B is an adaptor protein found at the interface of membrane and the cytoskeleton. Previous studies demonstrated that 4.1B serves as tumor suppressor. RESULTS We showed that 4.1B expression was decreased or lost in most GC patients. The expression pattern of it was tightly correlated with tumor size, TNM stage and overall survival (OS). We further showed that 4.1B inhibited the proliferation of two GC cell lines, MGC-803 and MKN-45, by impeding the EGFR/MAPK/ERK1/2 and PI3K/AKT pathways. A similar phenotype was also observed in immortalized mouse embryonic fibroblasts (MEF) derived from wild type (WT) and 4.1B knock-out (BKO) mice. Additionally, immunofluorescence (IF) staining and Co-IP showed that protein 4.1B bound to EGFR. Furthermore, the FERM domain of 4.1B interacted with EGFR through the initial 13 amino acids (P13) of the intracellular juxtamembrane (JM) segment of EGFR. The binding of 4.1B to EGFR inhibited dimerization and autophosphorylation of EGFR. CONCLUSION Our present work revealed that 4.1B plays important regulatory roles in the proliferation of GC cells by binding to EGFR and inhibiting EGFR function through an EGFR/MAPK/ERK1/2 pathway. Our results provide novel insight into the mechanism of the development and progression of GC.
Collapse
Affiliation(s)
- Fumin Xue
- Department of Gastroenterology, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
- Key Laboratory of H. pylori and Gastrointestinal Microecology of Henan Province, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Chao An
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Lixiang Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan China
| | - Gang Liu
- Department of Public Health, Zibo Integrate traditional Chinese & Western Medicine Hospital, Zibo, 255000 Shandong China
| | - Feifei Ren
- Key Laboratory of H. pylori and Gastrointestinal Microecology of Henan Province, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Xinhua Guo
- Red Cell Physiology, New York Blood Center, New York, NY 10065 USA
| | - Haibin Sun
- Department of Pathology, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Lu Mei
- Department of Gastroenterology, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Xiangdong Sun
- Key Laboratory of H. pylori and Gastrointestinal Microecology of Henan Province, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Jinpeng Li
- Department of Gastroenterology, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Youcai Tang
- Department of Pediatrics, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065 USA
| | - Pengyuan Zheng
- Department of Gastroenterology, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| |
Collapse
|
18
|
Xu F, Si X, Du J, Xu F, Yang A, Zhang C, Zhang X, Yang Y. Downregulating SynCAM and MPP6 expression is associated with ovarian cancer progression. Oncol Lett 2019; 18:2477-2483. [PMID: 31402947 PMCID: PMC6676726 DOI: 10.3892/ol.2019.10542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 05/17/2019] [Indexed: 12/13/2022] Open
Abstract
Synaptic cell adhesion molecules (SynCAMs) are single transmembrane proteins that belong to the immunoglobulin superfamily of cell adhesion molecules. In the present study, a decrease in SynCAM levels in ovarian tumor tissues compared with normal tissues is reported; the downregulation was accompanied by the grade malignancy. The observations suggested that SynCAM may be essential for important novel functions in ovarian cancer. Further experiments showed that low SynCAM expression inhibited membrane palmitoylated protein 6 (MPP6) expression, a member of the palmitoylated membrane protein subfamily of peripheral membrane-associated guanylate kinases. In addition, low levels of MPP6 in ovarian tumor tissues correlated with shorter patient survival. A SynCAM-regulated pathway may provide molecular targets for the treatment of ovarian cancer and novel biomarkers to be used in clinical diagnosis.
Collapse
Affiliation(s)
- Feixue Xu
- Department of Gynecology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xiaoqiang Si
- Department of Plastic Surgery, The Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Jingran Du
- Department of Gynecology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Feihua Xu
- Department of Labor and Environmental Health, The School of Public Health of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Aihong Yang
- Department of Gynecology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Caixia Zhang
- Department of Gynecology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xiucai Zhang
- Department of Gynecology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yongxiu Yang
- Department of Gynecology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
19
|
Abstract
Fibrosis is a medical condition characterized by an excessive deposition of extracellular matrix compounds such as collagen in tissues. Fibrotic lesions are present in many diseases and can affect all organs. The excessive extracellular matrix accumulation in these conditions can often have serious consequences and in many cases be life-threatening. A typical event seen in many fibrotic conditions is a profound accumulation of mast cells (MCs), suggesting that these cells can contribute to the pathology. Indeed, there is now substantialv evidence pointing to an important role of MCs in fibrotic disease. However, investigations from various clinical settings and different animal models have arrived at partly contradictory conclusions as to how MCs affect fibrosis, with many studies suggesting a detrimental role of MCs whereas others suggest that MCs can be protective. Here, we review the current knowledge of how MCs can affect fibrosis.
Collapse
Affiliation(s)
- Peter Bradding
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester, UK
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.,Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
20
|
Oncogenic role of SFRP2 in p53-mutant osteosarcoma development via autocrine and paracrine mechanism. Proc Natl Acad Sci U S A 2018; 115:E11128-E11137. [PMID: 30385632 DOI: 10.1073/pnas.1814044115] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Osteosarcoma (OS), the most common primary bone tumor, is highly metastatic with high chemotherapeutic resistance and poor survival rates. Using induced pluripotent stem cells (iPSCs) generated from Li-Fraumeni syndrome (LFS) patients, we investigate an oncogenic role of secreted frizzled-related protein 2 (SFRP2) in p53 mutation-associated OS development. Interestingly, we find that high SFRP2 expression in OS patient samples correlates with poor survival. Systems-level analyses identified that expression of SFRP2 increases during LFS OS development and can induce angiogenesis. Ectopic SFRP2 overexpression in normal osteoblast precursors is sufficient to suppress normal osteoblast differentiation and to promote OS phenotypes through induction of oncogenic molecules such as FOXM1 and CYR61 in a β-catenin-independent manner. Conversely, inhibition of SFRP2, FOXM1, or CYR61 represses the tumorigenic potential. In summary, these findings demonstrate the oncogenic role of SFRP2 in the development of p53 mutation-associated OS and that inhibition of SFRP2 is a potential therapeutic strategy.
Collapse
|
21
|
Kimura R, Yoneshige A, Hagiyama M, Otani T, Inoue T, Shiraishi N, Yanagihara K, Wakayama T, Ito A. Expression of cell adhesion molecule 1 in gastric neck and base glandular cells: Possible involvement in peritoneal dissemination of signet ring cells. Life Sci 2018; 213:206-213. [PMID: 30312702 DOI: 10.1016/j.lfs.2018.10.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 09/22/2018] [Accepted: 10/08/2018] [Indexed: 01/27/2023]
Abstract
AIMS To determine cellular distribution of cell adhesion molecule 1 (CADM1), an immunoglobulin superfamily member, in the human oxyntic gastric mucosa, and to explore possible involvement in the development and peritoneal dissemination of signet ring cell (SRC) gastric carcinoma, which often develops in the oxyntic mucosa. MAIN METHODS Immunohistochemistry and double immunofluorescence were conducted on surgical specimens of normal and SRC-bearing stomachs and peritoneal metastatic foci of SRCs. KATO-III (lacking CADM1) and HSC-43 (expressing CADM1) SRC cell lines were cocultured on a Met-5A mesothelial or TIG-1 fibroblastic cell monolayer. KEY FINDINGS In the oxyntic gland, some neck and nearly all base glandular cells were CADM1-positive, and mucin 5AC-positive cells were CADM1-negative, while some mucin 6-positive neck cells were CADM1-positive. Foveolar-epithelial, parietal, and endocrine cells were CADM1-negative. CADM1 was negative in all SRC carcinomas that were confined within the submucosa (n = 11) and all but one of those invading deeper (n = 15). In contrast, peritoneal metastatic foci of SRCs were CADM1-positive in five out of eleven cases (P < 0.01). In the cocultures, exogenous CADM1 made KATO-III cells adhere more and grow faster on a Met-5A monolayer, not on TIG-1 monolayers. HSC-43 cells adhered more and grew faster on Met-5A than on TIG-1 monolayers, which were partly counteracted by a function-neutralizing anti-CADM1 antibody. SIGNIFICANCE Nearly all chief cells and a part of mucous neck cells express CADM1. SRC gastric carcinoma appears to emerge as a CADM1-negative tumor, but CADM1 may help SRCs develop peritoneal dissemination through promoting their adhesion and growth in the serosal tissue.
Collapse
Affiliation(s)
- Ryuichiro Kimura
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Azusa Yoneshige
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Man Hagiyama
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Tomoyuki Otani
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Takao Inoue
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Naoki Shiraishi
- Hospital Pathology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Kazuyoshi Yanagihara
- Division of Biomarker Discovery, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Tomohiko Wakayama
- Department of Histology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Akihiko Ito
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan.
| |
Collapse
|
22
|
Zhang Y, Wang LJ, Yang HQ, Wang R, Wu HJ. MicroRNA-10b expression predicts long-term survival in patients with solid tumor. J Cell Physiol 2018; 234:1248-1256. [PMID: 30191959 DOI: 10.1002/jcp.27138] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 07/09/2018] [Indexed: 01/23/2023]
Abstract
BACKGROUND Numerous studies have evaluated the significance of the microRNA-10b (miR-10b) in the development and progression of many cancers. Their findings revealed that increased expression of miR-10b is associated with unfavorable prognosis in patients with cancer. RESULTS A total of 1,834 patients from 19 studies were included in this study. A significantly shorter overall survival was observed in patients with increased expression of miR-10b (hazard ratio [HR] = 1.99, 95% confidence interval [CI]: 1.51-2.61). Statistical significance was also observed in subgroup meta-analysis stratified by the cancer type, cutoff value, analysis type, and sample size. Also, patients with a high expression level of miR-10b had a poorer disease-free survival rate (HR = 1.18, 95% CI: 1.05-1.33). In addition, the pooled odds ratios (ORs) showed that increased miR-10b was also associated with positive lymph node metastasis (OR = 2.09, 95% CI: 1.45-3.03), distant metastasis (OR = 2.40, 95% CI: 1.57-3.67), tumor size (OR = 3.86, 95% CI: 2.25-6.64), and poor clinical stage (OR = 5.02, 95% CI: 3.37-7.47). MATERIALS AND METHODS A systematic literature search was conducted on a number of electronic databases, including PubMed, Embase, Web of Science, China National Knowledge Infrastructure, Springer, Google Scholar, and Gene expression omnibus. We retrieved the relevant articles to examine the association between the miR-10b expression levels and patients' prognosis. The meta-analysis was conducted using the RevMan 5.2 software and Stata SE12.0 software. CONCLUSIONS High miR-10b expression was correlated with poor clinical outcome, which indicated the potential clinical use of miR-10b as a molecular biomarker for cancer, particularly in assessing prognosis for patients with cancers. Further studies should be performed to verify the clinical utility of miR-10b in human solid tumors.
Collapse
Affiliation(s)
- Yi Zhang
- Department of General Surgery, The First People's Hospital of Neijiang, Neijiang, Sichuan, China
| | - Li-Juan Wang
- Department of Nephrology, ShangRao People's Hospital, ShangRao, Jiangxi, China
| | - He-Quan Yang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Rong Wang
- Department of General Surgery, The First People's Hospital of Neijiang, Neijiang, Sichuan, China
| | - Hua-Jun Wu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
23
|
Pan T, Hu X, Liu T, Xu Z, Wan N, Zhang Y, Li S. MiR-128-1-5p regulates tight junction induced by selenium deficiency via targeting cell adhesion molecule 1 in broilers vein endothelial cells. J Cell Physiol 2018; 233:8802-8814. [PMID: 29904913 DOI: 10.1002/jcp.26794] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/30/2018] [Indexed: 01/05/2023]
Abstract
Vein endothelial cells (VECs) constitute an important barrier for macromolecules and circulating cells from the blood to the tissues, stabilizing the colloid osmotic pressure of the blood, regulating the vascular tone, and rapidly changing the intercellular connection, and maintaining normal physiological function. Tight junction has been discovered as an important structural basis of intercellular connection and may play a key role in intercellular connection injuries or vascular diseases and selenium (Se) deficiency symptoms. Hence, we replicated the Se-deficient broilers model and detected the specific microRNA in response to Se-deficient vein by using quantitative real time-PCR (qRT-PCR) analysis. Also, we selected miR-128-1-5p based on differential expression in vein tissue and confirmed its target gene cell adhesion molecule 1 (CADM1) by the dual luciferase reporter assay and qRT-PCR in VECs. We made the ectopic miR-128-1-5p expression for the purpose of validating its function on tight junction. The result showed that miR-128-1-5p and CADM1 were involved in the ZO-1-mediated tight junction, increased paracellular permeability, and arrested cell cycle. We presumed that miR-128-1-5p and Se deficiency might trigger tight junction. Interestingly, miR-128-1-5p inhibitor and fasudil in part hinder the destruction of the intercellular structure caused by Se deficiency. The miR-128-1-5p/CADM1/tight junction axis provides a new avenue toward understanding the mechanism of Se deficiency, revealing a novel regulation model of tight junction injury in vascular diseases.
Collapse
Affiliation(s)
- Tingru Pan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xueyuan Hu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Tianqi Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhe Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Na Wan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yiming Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
24
|
Zhang Y, Liao RB, Hu LL, Tong BX, Hao TF, Wu HJ. The microRNA miR-10b as a potentially promising biomarker to predict the prognosis of cancer patients: a meta-analysis. Oncotarget 2017; 8:104543-104551. [PMID: 29262659 PMCID: PMC5732825 DOI: 10.18632/oncotarget.21428] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/17/2017] [Indexed: 01/04/2023] Open
Abstract
Background Reported studies on carcinoma have evaluated the significance of the microRNA miR-10b in the development and progression of many cancers. Increased expression of miR-10b is associated with the susceptibility to lymph node metastasis and distant metastasis in various tumors. Results The results of the meta-analysis revealed that lymph node metastasis occurred more frequently in the patients group with high expression level of miR-10b than in the patients group with low expression level of miR-10b (OR=4.65, 95% CI: 3.40-6.37, P <0.00001, fixed-effects model). Additionally, a similar result was observed in the association between miR-10b expression and distant metastasis (OR=2.70, 95% CI: 1.79-4.08, P <0.00001, fixed-effects model). Materials and Methods In this study, a meta-analysis, including the majority of the relevant articles, was conducted to investigate the association of the miR-10b expression level with metastasis in cancer patients. Systematic literature retrieval was performed by searching in a number of electronic databases. The meta-analysis was conducted using the RevMan 5.2 software and Stata SE12.0 software. A total of 962 patients with carcinoma from 9 studies were included in analysis. Conclusions This meta-analysis demonstrated that the overexpression of miR-10b was significantly correlated with metastasis status, and indicated the potential clinical use of miR-10b as a molecular biomarker, particularly in assessing prognosis for patients with cancers.
Collapse
Affiliation(s)
- Yi Zhang
- Department of General Surgery, The First People's Hospital of Neijiang, Neijiang 641000, Sichuan Province, P.R. China
| | - Rong-Bo Liao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, P.R. China
| | - Li-Lin Hu
- Department of Medicine, Nanchang University, Nanchang 330000, Jiangxi Province, P.R. China
| | - Bi-Xia Tong
- Department of Nursing, Shangrao People's Hospital, Shangrao 334000, Jiangxi Province, P.R. China
| | - Teng-Fei Hao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, P.R. China
| | - Hua-Jun Wu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, P.R. China
| |
Collapse
|
25
|
Rademacher N, Schmerl B, Lardong JA, Wahl MC, Shoichet SA. MPP2 is a postsynaptic MAGUK scaffold protein that links SynCAM1 cell adhesion molecules to core components of the postsynaptic density. Sci Rep 2016; 6:35283. [PMID: 27756895 PMCID: PMC5069480 DOI: 10.1038/srep35283] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/15/2016] [Indexed: 02/07/2023] Open
Abstract
At neuronal synapses, multiprotein complexes of trans-synaptic adhesion molecules, scaffold proteins and neurotransmitter receptors assemble to essential building blocks required for synapse formation and maintenance. Here we describe a novel role for the membrane-associated guanylate kinase (MAGUK) protein MPP2 (MAGUK p55 subfamily member 2) at synapses of rat central neurons. Through interactions mediated by its C-terminal SH3-GK domain module, MPP2 binds to the abundant postsynaptic scaffold proteins PSD-95 and GKAP and localises to postsynaptic sites in hippocampal neurons. MPP2 also colocalises with the synaptic adhesion molecule SynCAM1. We demonstrate that the SynCAM1 C-terminus interacts directly with the MPP2 PDZ domain and that MPP2 does not interact in this manner with other highly abundant postsynaptic transmembrane proteins. Our results highlight a previously unexplored role for MPP2 at postsynaptic sites as a scaffold that links SynCAM1 cell adhesion molecules to core proteins of the postsynaptic density.
Collapse
Affiliation(s)
- Nils Rademacher
- Neuroscience Research Center and Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Bettina Schmerl
- Neuroscience Research Center and Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jennifer A. Lardong
- Institute of Chemistry and Biochemistry, Structural Biochemistry, Freie Universität Berlin, Takustr. 6, 14195 Berlin, Germany
| | - Markus C. Wahl
- Institute of Chemistry and Biochemistry, Structural Biochemistry, Freie Universität Berlin, Takustr. 6, 14195 Berlin, Germany
| | - Sarah A. Shoichet
- Neuroscience Research Center and Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
26
|
Abstract
Mast cells (MCs) play a central role in tissue homoeostasis, sensing the local environment through numerous innate cell surface receptors. This enables them to respond rapidly to perceived tissue insults with a view to initiating a co-ordinated programme of inflammation and repair. However, when the tissue insult is chronic, the ongoing release of multiple pro-inflammatory mediators, proteases, cytokines and chemokines leads to tissue damage and remodelling. In asthma, there is strong evidence of ongoing MC activation, and their mediators and cell-cell signals are capable of regulating many facets of asthma pathophysiology. This article reviews the evidence behind this.
Collapse
Affiliation(s)
- P Bradding
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester, UK
| | - G Arthur
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester, UK
| |
Collapse
|
27
|
Zhu Q, Gong L, Wang J, Tu Q, Yao L, Zhang JR, Han XJ, Zhu SJ, Wang SM, Li YH, Zhang W. miR-10b exerts oncogenic activity in human hepatocellular carcinoma cells by targeting expression of CUB and sushi multiple domains 1 (CSMD1). BMC Cancer 2016; 16:806. [PMID: 27756250 PMCID: PMC5069781 DOI: 10.1186/s12885-016-2801-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 09/22/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a lethal disease, while the precise underlying molecular mechanisms of HCC pathogenesis remain to be defined. MicroRNA (miRNA), a class of non-coding small RNAs, can post-transcriptionally regulate gene expression. Altered miRNA expression has been reported in HCCs. This study assessed expression and the oncogenic activity of miRNA-10b (miR-10b) in HCC. METHODS Forty-five paired human HCC and adjacent non-tumor tissues were collected for qRT-PCR and immunohistochemistry analysis of miR-10b and CUB and Sushi multiple domains 1 (CSMD1), respectively. We analyzed the clinicopathological data from these patients to further determine if there was an association between miR-10b and CSMD1. HCC cell lines were used to assess the effects of miR-10b mimics or inhibitors on cell viability, migration, invasion, cell cycle distribution, and colony formation. Luciferase assay was used to assess miR-10b binding to the 3'-untranslated region (3'-UTR) of CSMD1. RESULTS miR-10b was highly expressed in HCC tissues compared to normal tissues. In vitro, overexpression of miR-10b enhanced HCC cell viability, migration, and invasion; whereas, downregulation of miR-10b expression suppressed these properties in HCC cells. Injection of miR-10b mimics into tumor cell xenografts also promoted xenograft growth in nude mice. Bioinformatics and luciferase reporter assay demonstrated that CSMD1 was the target gene of miR-10b. Immunocytochemical, immunohistochemical, and qRT-PCR data indicated that miR-10b decreased CSMD1 expression in HCC cells. CONCLUSIONS We showed that miR-10b is overexpressed in HCC tissues and miR-10b mimics promoted HCC cell viability and invasion via targeting CSMD1 expression. Our findings suggest that miR-10b acts as an oncogene by targeting the tumor suppressor gene, CSMD1, in HCC.
Collapse
Affiliation(s)
- Qiao Zhu
- The Helmholtz Sino-German Laboratory for Cancer Research, Department of Pathology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Li Gong
- The Helmholtz Sino-German Laboratory for Cancer Research, Department of Pathology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Jun Wang
- The Helmholtz Sino-German Laboratory for Cancer Research, Department of Pathology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Qian Tu
- The Helmholtz Sino-German Laboratory for Cancer Research, Department of Pathology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Li Yao
- The Helmholtz Sino-German Laboratory for Cancer Research, Department of Pathology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Jia-Rui Zhang
- The Helmholtz Sino-German Laboratory for Cancer Research, Department of Pathology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Xiu-Juan Han
- The Helmholtz Sino-German Laboratory for Cancer Research, Department of Pathology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Shao-Jun Zhu
- The Helmholtz Sino-German Laboratory for Cancer Research, Department of Pathology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Shu-Mei Wang
- The Helmholtz Sino-German Laboratory for Cancer Research, Department of Pathology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Yan-Hong Li
- The Helmholtz Sino-German Laboratory for Cancer Research, Department of Pathology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China. .,Department of Gynecology and Obstetrics, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China. .,Department of Pathology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.
| | - Wei Zhang
- The Helmholtz Sino-German Laboratory for Cancer Research, Department of Pathology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China. .,Department of Pathology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.
| |
Collapse
|
28
|
Zhang C, Caldwell TA, Mirbolooki MR, Duong D, Park EJ, Chi NW, Chessler SD. Extracellular CADM1 interactions influence insulin secretion by rat and human islet β-cells and promote clustering of syntaxin-1. Am J Physiol Endocrinol Metab 2016; 310:E874-85. [PMID: 27072493 PMCID: PMC4935136 DOI: 10.1152/ajpendo.00318.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 04/08/2016] [Indexed: 11/22/2022]
Abstract
Contact between β-cells is necessary for their normal function. Identification of the proteins mediating the effects of β-cell-to-β-cell contact is a necessary step toward gaining a full understanding of the determinants of β-cell function and insulin secretion. The secretory machinery of the β-cells is nearly identical to that of central nervous system (CNS) synapses, and we hypothesize that the transcellular protein interactions that drive maturation of the two secretory machineries upon contact of one cell (or neural process) with another are also highly similar. Two such transcellular interactions, important for both synaptic and β-cell function, have been identified: EphA/ephrin-A and neuroligin/neurexin. Here, we tested the role of another synaptic cleft protein, CADM1, in insulinoma cells and in rat and human islet β-cells. We found that CADM1 is a predominant CADM isoform in β-cells. In INS-1 cells and primary β-cells, CADM1 constrains insulin secretion, and its expression decreases after prolonged glucose stimulation. Using a coculture model, we found that CADM1 also influences insulin secretion in a transcellular manner. We asked whether extracellular CADM1 interactions exert their influence via the same mechanisms by which they influence neurotransmitter exocytosis. Our results suggest that, as in the CNS, CADM1 interactions drive exocytic site assembly and promote actin network formation. These results support the broader hypothesis that the effects of cell-cell contact on β-cell maturation and function are mediated by the same extracellular protein interactions that drive the formation of the presynaptic exocytic machinery. These interactions may be therapeutic targets for reversing β-cell dysfunction in diabetes.
Collapse
Affiliation(s)
- Charles Zhang
- Department of Medicine, University of California, Irvine, School of Medicine, Irvine, California
| | - Thomas A Caldwell
- Department of Medicine, University of California, Irvine, School of Medicine, Irvine, California
| | - M Reza Mirbolooki
- Department of Medicine, University of California, Irvine, School of Medicine, Irvine, California
| | - Diana Duong
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, California; and
| | - Eun Jee Park
- Department of Medicine, University of California, Irvine, School of Medicine, Irvine, California
| | - Nai-Wen Chi
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Steven D Chessler
- Department of Medicine, University of California, Irvine, School of Medicine, Irvine, California;
| |
Collapse
|
29
|
Iwasaki T, Matsushita M, Nonaka D, Nagata K, Kato M, Kuwamoto S, Murakami I, Hayashi K. Lower expression of CADM1 and higher expression of MAL in Merkel cell carcinomas are associated with Merkel cell polyomavirus infection and better prognosis. Hum Pathol 2016; 48:1-8. [DOI: 10.1016/j.humpath.2015.09.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 09/06/2015] [Accepted: 09/23/2015] [Indexed: 11/15/2022]
|
30
|
Perez-Janices N, Blanco-Luquin I, Tuñón MT, Barba-Ramos E, Ibáñez B, Zazpe-Cenoz I, Martinez-Aguillo M, Hernandez B, Martínez-Lopez E, Fernández AF, Mercado MR, Cabada T, Escors D, Megias D, Guerrero-Setas D. EPB41L3, TSP-1 and RASSF2 as new clinically relevant prognostic biomarkers in diffuse gliomas. Oncotarget 2016; 6:368-80. [PMID: 25621889 PMCID: PMC4381601 DOI: 10.18632/oncotarget.2745] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 11/12/2014] [Indexed: 12/17/2022] Open
Abstract
Hypermethylation of tumor suppressor genes is one of the hallmarks in the progression of brain tumors. Our objectives were to analyze the presence of the hypermethylation of EPB41L3, RASSF2 and TSP-1 genes in 132 diffuse gliomas (astrocytic and oligodendroglial tumors) and in 10 cases of normal brain, and to establish their association with the patients’ clinicopathological characteristics. Gene hypermethylation was analyzed by methylation-specific-PCR and confirmed by pyrosequencing (for EPB41L3 and TSP-1) and bisulfite-sequencing (for RASSF2). EPB41L3, RASSF2 and TSP-1 genes were hypermethylated only in tumors (29%, 10.6%, and 50%, respectively), confirming their cancer-specific role. Treatment of cells with the DNA-demethylating-agent 5-aza-2′-deoxycytidine restores their transcription, as confirmed by quantitative-reverse-transcription-PCR and immunofluorescence. Immunohistochemistry for EPB41L3, RASSF2 and TSP-1 was performed to analyze protein expression; p53, ki-67, and CD31 expression and 1p/19q co-deletion were considered to better characterize the tumors. EPB41L3 and TSP-1 hypermethylation was associated with worse (p = 0.047) and better (p = 0.037) prognosis, respectively. This observation was confirmed after adjusting the results for age and tumor grade, the role of TSP-1 being most pronounced in oligodendrogliomas (p = 0.001). We conclude that EPB41L3, RASSF2 and TSP-1 genes are involved in the pathogenesis of diffuse gliomas, and that EPB41L3 and TSP-1 hypermethylation are of prognostic significance.
Collapse
Affiliation(s)
- N Perez-Janices
- Cancer Epigenetics Group, Navarrabiomed-Fundación Miguel Servet, Navarra, Spain
| | - I Blanco-Luquin
- Cancer Epigenetics Group, Navarrabiomed-Fundación Miguel Servet, Navarra, Spain
| | - M T Tuñón
- Department of Pathology Section A, Complejo Hospitalario de Navarra, Navarra Health Service, Navarra, Spain
| | - E Barba-Ramos
- Department of Pathology Section A, Complejo Hospitalario de Navarra, Navarra Health Service, Navarra, Spain
| | - B Ibáñez
- Navarrabiomed-Fundación Miguel Servet, Navarra, Spain. Red de Evaluación en Servicios Sanitarios y Enfermedades Crónicas (REDISSEC), Navarra, Spain
| | - I Zazpe-Cenoz
- Department of Neurosurgery, Complejo Hospitalario de Navarra, Navarra Health Service, Navarra, Spain
| | - M Martinez-Aguillo
- Department of Medical Oncology, Complejo Hospitalario de Navarra, Navarra Health Service, Navarra, Spain
| | - B Hernandez
- Department of Medical Oncology, Complejo Hospitalario de Navarra, Navarra Health Service, Navarra, Spain
| | - E Martínez-Lopez
- Department of Radiation Oncology, Complejo Hospitalario de Navarra, Navarra Health Service, Navarra, Spain
| | - A F Fernández
- Cancer Epigenetics Laboratory, Instituto Universitario de Oncología del Principado de Asturias (IUOPA), HUCA, Universidad de Oviedo, Asturias, Spain
| | - M R Mercado
- Department of Pathology Section A, Complejo Hospitalario de Navarra, Navarra Health Service, Navarra, Spain
| | - T Cabada
- Department of Radiology, Complejo Hospitalario de Navarra, Navarra Health Service, Navarra, Spain
| | - D Escors
- Navarrabiomed-Fundación Miguel Servet, Navarra, Spain
| | - D Megias
- Confocal Microscopy Core Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - D Guerrero-Setas
- Cancer Epigenetics Group, Navarrabiomed-Fundación Miguel Servet, Navarra, Spain
| |
Collapse
|
31
|
Yoneshige A, Hagiyama M, Fujita M, Ito A. Pathogenic Actions of Cell Adhesion Molecule 1 in Pulmonary Emphysema and Atopic Dermatitis. Front Cell Dev Biol 2015; 3:75. [PMID: 26636084 PMCID: PMC4653308 DOI: 10.3389/fcell.2015.00075] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 11/09/2015] [Indexed: 12/27/2022] Open
Abstract
Cell adhesion mediated by adhesion molecules is of central importance in the maintenance of tissue homeostasis. Therefore, altered expression of adhesion molecules leads to the development of various tissue disorders involving cell activation, degeneration, and apoptosis. Nevertheless, it still remains unclear what initiates the altered expression of adhesion molecules and how the subsequent pathological cascades proceed. In this regard, cell adhesion molecule 1 (CADM1) is one of the candidates that is involved in the development of pathological lesions; it is an intercellular adhesion molecule that is expressed in various types of cells such as pulmonary cells, neurons, and mast cells. Recent studies have revealed that alterations in the transcriptional or post-transcriptional expressions of CADM1 correlate with the pathogenesis of pulmonary diseases and allergic diseases. In this review, we specifically focus on how CADM1 is involved in the development of pathological lesions in pulmonary emphysema and atopic dermatitis.
Collapse
Affiliation(s)
- Azusa Yoneshige
- Department of Pathology, Faculty of Medicine, Kinki University Osaka, Japan
| | - Man Hagiyama
- Department of Pathology, Faculty of Medicine, Kinki University Osaka, Japan
| | - Mitsugu Fujita
- Department of Microbiology, Faculty of Medicine, Kinki University Osaka, Japan
| | - Akihiko Ito
- Department of Pathology, Faculty of Medicine, Kinki University Osaka, Japan
| |
Collapse
|
32
|
Yoneshige A, Hagiyama M, Inoue T, Mimae T, Kato T, Okada M, Enoki E, Ito A. Increased ectodomain shedding of cell adhesion molecule 1 as a cause of type II alveolar epithelial cell apoptosis in patients with idiopathic interstitial pneumonia. Respir Res 2015; 16:90. [PMID: 26231557 PMCID: PMC4531801 DOI: 10.1186/s12931-015-0255-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 07/21/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Lung alveolar epithelial cell (AEC) apoptosis has attracted attention as an early pathogenic event in the development of idiopathic interstitial pneumonia (IIP); however, the causative mechanism remains unclear. Cell adhesion molecule 1 (CADM1) is an AEC adhesion molecule in the immunoglobulin superfamily. It generates a membrane-associated C-terminal fragment, αCTF, through protease-mediated ectodomain shedding, termed α-shedding. Increased CADM1 α-shedding contributes to AEC apoptosis in emphysematous lungs. METHODS Formalin-fixed, paraffin-embedded lung lobes (n = 39) from 36 autopsied patients with IIP were classified as acute IIP (n = 10), fibrosing-type nonspecific IIP (f-NSIP, n = 10), cryptogenic organizing IIP (n = 9), and usual IIP (n = 10). CADM1 expression in the lung sections was examined by western blotting and compared with control lungs (n = 10). The rate of CADM1 α-shedding was calculated as the relative amount of αCTF to full-length CADM1, and the full-length CADM1 level was estimated per epithelial cell by normalization to cytokeratin 7, a lung epithelial marker. Apoptotic AECs were detected by immunohistochemistry for single-stranded DNA (ssDNA). NCI-H441 and A549 human lung epithelial cells were transfected with small interfering RNA (siRNA) to silence CADM1 expression and analyzed by terminal nucleotide nick end labeling assays. RESULTS The rate of CADM1 α-shedding was higher in all IIP subtypes than in the control (P ≤ 0.019), and the full-length CADM1 level was lower in f-NSIP (P = 0.007). The α-shedding rate and full-length CADM1 level were correlated with each other (P = 0.015) and with the proportion of ssDNA-positive AECs (P ≤ 0.024). NCI-H441 cells transfected with siRNA exhibited a 61 % lower rate of expression of full-length CADM1 and a 17-fold increased proportion of apoptotic cells. Similar results were obtained with A549 cells. CONCLUSIONS CADM1 α-shedding appeared to be increased in all four IIP subtypes and consequently contributed to AEC apoptosis by decreasing the full-length CADM1 level. This mechanism particularly impacted f-NSIP. The molecular mechanism causing AEC apoptosis may be similar between IIP and emphysema.
Collapse
Affiliation(s)
- Azusa Yoneshige
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka, 589-8511, Japan
| | - Man Hagiyama
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka, 589-8511, Japan
| | - Takao Inoue
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka, 589-8511, Japan
| | - Takahiro Mimae
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Takashi Kato
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka, 589-8511, Japan
| | - Morihito Okada
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Eisuke Enoki
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka, 589-8511, Japan
| | - Akihiko Ito
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka, 589-8511, Japan.
| |
Collapse
|
33
|
Sakurai-Yageta M, Maruyama T, Suzuki T, Ichikawa K, Murakami Y. Dynamic regulation of a cell adhesion protein complex including CADM1 by combinatorial analysis of FRAP with exponential curve-fitting. PLoS One 2015; 10:e0116637. [PMID: 25780926 PMCID: PMC4364555 DOI: 10.1371/journal.pone.0116637] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 12/11/2014] [Indexed: 11/19/2022] Open
Abstract
Protein components of cell adhesion machinery show continuous renewal even in the static state of epithelial cells and participate in the formation and maintenance of normal epithelial architecture and tumor suppression. CADM1 is a tumor suppressor belonging to the immunoglobulin superfamily of cell adhesion molecule and forms a cell adhesion complex with an actin-binding protein, 4.1B, and a scaffold protein, MPP3, in the cytoplasm. Here, we investigate dynamic regulation of the CADM1-4.1B-MPP3 complex in mature cell adhesion by fluorescence recovery after photobleaching (FRAP) analysis. Traditional FRAP analysis were performed for relatively short period of around 10 min. Here, thanks to recent advances in the sensitive laser detector systems, we examine FRAP of CADM1 complex for longer period of 60 min and analyze the recovery with exponential curve-fitting to distinguish the fractions with different diffusion constants. This approach reveals that the fluorescence recovery of CADM1 is fitted to a single exponential function with a time constant (τ) of approximately 16 min, whereas 4.1B and MPP3 are fitted to a double exponential function with two τs of approximately 40-60 sec and 16 min. The longer τ is similar to that of CADM1, suggesting that 4.1B and MPP3 have two distinct fractions, one forming a complex with CADM1 and the other present as a free pool. Fluorescence loss in photobleaching analysis supports the presence of a free pool of these proteins near the plasma membrane. Furthermore, double exponential fitting makes it possible to estimate the ratio of 4.1B and MPP3 present as a free pool and as a complex with CADM1 as approximately 3:2 and 3:1, respectively. Our analyses reveal a central role of CADM1 in stabilizing the complex with 4.1B and MPP3 and provide insight in the dynamics of adhesion complex formation.
Collapse
Affiliation(s)
- Mika Sakurai-Yageta
- Division of Molecular Pathology, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Tomoko Maruyama
- Division of Molecular Pathology, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Takashi Suzuki
- Japan Science and Technology Agency, CREST, 4-5-3, Yonbancho, Chiyoda-ku, Tokyo, 102-8666, Japan
- The Division of Mathematical Science, Graduate School of Engineering Science, Osaka University, 1-3, Machikaneyama-cho, Toyonaka, Osaka, 560-8531, Japan
| | - Kazuhisa Ichikawa
- Division of Mathematical Oncology, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
- Japan Science and Technology Agency, CREST, 4-5-3, Yonbancho, Chiyoda-ku, Tokyo, 102-8666, Japan
| | - Yoshinori Murakami
- Division of Molecular Pathology, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
- Japan Science and Technology Agency, CREST, 4-5-3, Yonbancho, Chiyoda-ku, Tokyo, 102-8666, Japan
- * E-mail:
| |
Collapse
|
34
|
de Haan HG, Bezemer ID, Vossen CY, van Hylckama Vlieg A, Böehringer S, Hasstedt SJ, Levy S, Rosendaal FR, Bovill EG. Genetic variants in Cell Adhesion Molecule 1 (CADM1): a validation study of a novel endothelial cell venous thrombosis risk factor. Thromb Res 2014; 134:1186-92. [PMID: 25306186 PMCID: PMC4252856 DOI: 10.1016/j.thromres.2014.09.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 08/28/2014] [Accepted: 09/09/2014] [Indexed: 12/21/2022]
Abstract
INTRODUCTION In a protein C deficient family, we recently identified a candidate gene, CADM1, which interacted with protein C deficiency in increasing the risk of venous thrombosis (VT). This study aimed to determine whether CADM1 variants also interact with protein C pathway abnormalities in increasing VT risk outside this family. MATERIALS AND METHODS We genotyped over 300 CADM1 variants in the population-based MEGA case-control study. We compared VT risks between cases with low protein C activity (n=194), low protein S levels (n=23), high factor VIII activity (n=165) or factor V Leiden carriers (n=580), and all 4004 controls. Positive associations were repeated in all 3496 cases and 4004 controls. RESULTS We found 22 variants which were associated with VT in one of the protein C pathway risk groups. After mutual adjustment, six variants remained associated with VT. The strongest evidence was found for rs220842 and rs11608105. For rs220842, the odds ratio (OR) for VT was 3.2 (95% CI 1.2-9.0) for cases with high factor VIII activity compared with controls. In addition, this variant was associated with an increased risk of VT in the overall study population (OR: 1.5, 95% CI 1.0-2.2). The other variant, rs11608105, was not associated with VT in the overall study population (OR: 1.0, 95% CI 0.8-1.1), but showed a strong effect on VT risk (OR: 21, 95% CI 5.1-88) when combined with low protein C or S levels. CONCLUSIONS In a population-based association study, we confirm a role for CADM1 variants in increasing the risk of VT by interaction with protein C pathway abnormalities.
Collapse
Affiliation(s)
- Hugoline G de Haan
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Irene D Bezemer
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Carla Y Vossen
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands; Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Stefan Böehringer
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Sandra J Hasstedt
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Samuel Levy
- Scripps Translational Science Institute, Scripps Research Institute, San Diego, CA, USA
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Edwin G Bovill
- Department of Pathology, University of Vermont, Burlington, VT, USA.
| |
Collapse
|
35
|
Wang Z, Zhang J, Ye M, Zhu M, Zhang B, Roy M, Liu J, An X. Tumor suppressor role of protein 4.1B/DAL-1. Cell Mol Life Sci 2014; 71:4815-30. [PMID: 25183197 PMCID: PMC11113756 DOI: 10.1007/s00018-014-1707-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 07/21/2014] [Accepted: 08/15/2014] [Indexed: 12/14/2022]
Abstract
Protein 4.1B/DAL-1 is a membrane skeletal protein that belongs to the protein 4.1 family. Protein 4.1B/DAL-1 is localized to sites of cell-cell contact and functions as an adapter protein, linking the plasma membrane to the cytoskeleton or associated cytoplasmic signaling effectors and facilitating their activities in various pathways. Protein 4.1B/DAL-1 is involved in various cytoskeleton-associated processes, such as cell motility and adhesion. Moreover, protein 4.1B/DAL-1 also plays a regulatory role in cell growth, differentiation, and the establishment of epithelial-like cell structures. Protein 4.1B/DAL-1 is normally expressed in multiple human tissues, but loss of its expression or prominent down-regulation of its expression is frequently observed in corresponding tumor tissues and tumor cell lines, suggesting that protein 4.1B/DAL-1 is involved in the molecular pathogenesis of these tumors and acts as a potential tumor suppressor. This review will focus on the structure of protein 4.1B/DAL-1, 4.1B/DAL-1-interacting molecules, 4.1B/DAL-1 inactivation and tumor progression, and anti-tumor activity of the 4.1B/DAL-1.
Collapse
Affiliation(s)
- Zi Wang
- Molecular Biology Research Center, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, 410078 China
| | - Ji Zhang
- Molecular Biology Research Center, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, 410078 China
- Department of Hematology, The First Affiliated Hospital, University of South China, Hengyang, 421001 China
| | - Mao Ye
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, 410082 China
| | - Min Zhu
- Molecular Biology Research Center, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, 410078 China
| | - Bin Zhang
- Department of Histology and Embryology, Xiangya School Medicine, Central South University, Changsha, 410083 China
| | - Mridul Roy
- Molecular Biology Research Center, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, 410078 China
| | - Jing Liu
- Molecular Biology Research Center, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, 410078 China
- State Key Laboratory of Medical Genetics, Central South University, 110 Xiangya Road, Changsha, 410078 China
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, 310 E 67th Street, New York, 10065 USA
| |
Collapse
|
36
|
Inoue T, Hagiyama M, Yoneshige A, Kato T, Enoki E, Maenishi O, Chikugo T, Kimura M, Satou T, Ito A. Increased ectodomain shedding of cell adhesion molecule 1 from pancreatic islets in type 2 diabetic pancreata: correlation with hemoglobin A1c levels. PLoS One 2014; 9:e100988. [PMID: 24964098 PMCID: PMC4071031 DOI: 10.1371/journal.pone.0100988] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 05/31/2014] [Indexed: 01/09/2023] Open
Abstract
Pulmonary emphysema and type 2 diabetes mellitus (T2DM), both caused by lifestyle factors, frequently concur. Respectively, the diseases affect lung alveolar and pancreatic islet cells, which express cell adhesion molecule 1 (CADM1), an immunoglobulin superfamily member. Protease-mediated ectodomain shedding of full-length CADM1 produces C-terminal fragments (CTFs) with proapoptotic activity. In emphysematous lungs, the CADM1 shedding rate and thus the level of CTFs in alveolar cells increase. In this study, CADM1 expression in islet cells was examined by western blotting. Protein was extracted from formalin-fixed, paraffin-embedded sections of pancreata isolated from patients with T2DM (n = 12) or from patients without pancreatic disease (n = 8) at autopsy. After adjusting for the number of islet cells present in the adjacent section, we found that full-length CADM1 decreased in T2DM islets, while ectodomain shedding increased. Hemoglobin A1c levels, measured when patients were alive, correlated inversely with full-length CADM1 levels (P = 0.041) and positively with ectodomain shedding rates (P = 0.001). In immunofluorescence images of T2DM islet cells, CADM1 was detected in the cytoplasm, but not on the cell membrane. Consistently, when MIN6-m9 mouse beta cells were treated with phorbol ester and trypsin to induce shedding, CADM1 immunostaining was diffuse in the cytoplasm. When a form of CTFs was exogenously expressed in MIN6-m9 cells, it localized diffusely in the cytoplasm and increased the number of apoptotic cells. These results suggest that increased CADM1 ectodomain shedding contributes to blood glucose dysregulation in T2DM by decreasing full-length CADM1 and producing CTFs that accumulate in the cytoplasm and promote apoptosis of beta cells. Thus, this study has identified a molecular alteration shared by pulmonary emphysema and T2DM.
Collapse
Affiliation(s)
- Takao Inoue
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka, Japan
| | - Man Hagiyama
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka, Japan
| | - Azusa Yoneshige
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka, Japan
| | - Takashi Kato
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka, Japan
| | - Eisuke Enoki
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka, Japan
| | - Osamu Maenishi
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka, Japan
| | - Takaaki Chikugo
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka, Japan
| | - Masatomo Kimura
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka, Japan
| | - Takao Satou
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka, Japan
| | - Akihiko Ito
- Department of Pathology, Faculty of Medicine, Kinki University, Osaka, Japan
- * E-mail:
| |
Collapse
|
37
|
Frei JA, Stoeckli ET. SynCAMs extend their functions beyond the synapse. Eur J Neurosci 2014; 39:1752-60. [DOI: 10.1111/ejn.12544] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 01/17/2014] [Accepted: 02/03/2014] [Indexed: 02/06/2023]
Affiliation(s)
- Jeannine A. Frei
- Institute of Molecular Life Sciences and Neuroscience Center Zurich; University of Zurich; Winterthurerstrasse 190 8057 Zurich Switzerland
| | - Esther T. Stoeckli
- Institute of Molecular Life Sciences and Neuroscience Center Zurich; University of Zurich; Winterthurerstrasse 190 8057 Zurich Switzerland
| |
Collapse
|
38
|
Murakami S, Sakurai-Yageta M, Maruyama T, Murakami Y. Trans-homophilic interaction of CADM1 activates PI3K by forming a complex with MAGuK-family proteins MPP3 and Dlg. PLoS One 2014; 9:e82894. [PMID: 24503895 PMCID: PMC3913574 DOI: 10.1371/journal.pone.0082894] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 11/07/2013] [Indexed: 11/25/2022] Open
Abstract
CADM1 (Cell adhesion molecule 1), a cell adhesion molecule belonging to the immunoglobulin superfamily, is involved in cell-cell interaction and the formation and maintenance of epithelial structure. Expression of CADM1 is frequently down-regulated in various tumors derived from epithelial cells. However, the intracellular signaling pathways activated by CADM1-mediated cell adhesion remain unknown. Here, we established a cell-based spreading assay to analyze the signaling pathway specifically activated by the trans-homophilic interaction of CADM1. In the assay, MDCK cells expressing exogenous CADM1 were incubated on the glass coated with a recombinant extracellular fragment of CADM1, and the degree of cell spreading was quantified by measuring their surface area. Assay screening of 104 chemical inhibitors with known functions revealed that LY294002, an inhibitor of phosphoinositide 3-kinase (PI3K), efficiently suppressed cell spreading in a dose-dependent manner. Inhibitors of Akt and Rac1, downstream effectors of PI3K, also partially suppressed cell spreading, while the addition of both inhibitors blocked cell spreading to the same extent as did LY294002. Furthermore, MPP3 and Dlg, membrane-associated guanylate kinase homologs (MAGuK) proteins, connect CADM1 with p85 of PI3K by forming a multi-protein complex at the periphery of cells. These results suggest that trans-homophilic interaction mediated by CADM1 activates the PI3K pathway to reorganize the actin cytoskeleton and form epithelial cell structure.
Collapse
Affiliation(s)
- Shigefumi Murakami
- Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Mika Sakurai-Yageta
- Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tomoko Maruyama
- Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoshinori Murakami
- Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
39
|
Moiseeva EP, Straatman KR, Leyland ML, Bradding P. CADM1 controls actin cytoskeleton assembly and regulates extracellular matrix adhesion in human mast cells. PLoS One 2014; 9:e85980. [PMID: 24465823 PMCID: PMC3899107 DOI: 10.1371/journal.pone.0085980] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 12/03/2013] [Indexed: 12/21/2022] Open
Abstract
CADM1 is a major receptor for the adhesion of mast cells (MCs) to fibroblasts, human airway smooth muscle cells (HASMCs) and neurons. It also regulates E-cadherin and alpha6beta4 integrin in other cell types. Here we investigated a role for CADM1 in MC adhesion to both cells and extracellular matrix (ECM). Downregulation of CADM1 in the human MC line HMC-1 resulted not only in reduced adhesion to HASMCs, but also reduced adhesion to their ECM. Time-course studies in the presence of EDTA to inhibit integrins demonstrated that CADM1 provided fast initial adhesion to HASMCs and assisted with slower adhesion to ECM. CADM1 downregulation, but not antibody-dependent CADM1 inhibition, reduced MC adhesion to ECM, suggesting indirect regulation of ECM adhesion. To investigate potential mechanisms, phosphotyrosine signalling and polymerisation of actin filaments, essential for integrin-mediated adhesion, were examined. Modulation of CADM1 expression positively correlated with surface KIT levels and polymerisation of cortical F-actin in HMC-1 cells. It also influenced phosphotyrosine signalling and KIT tyrosine autophosphorylation. CADM1 accounted for 46% of surface KIT levels and 31% of F-actin in HMC-1 cells. CADM1 downregulation resulted in elongation of cortical actin filaments in both HMC-1 cells and human lung MCs and increased cell rigidity of HMC-1 cells. Collectively these data suggest that CADM1 is a key adhesion receptor, which regulates MC net adhesion, both directly through CADM1-dependent adhesion, and indirectly through the regulation of other adhesion receptors. The latter is likely to occur via docking of KIT and polymerisation of cortical F-actin. Here we propose a stepwise model of adhesion with CADM1 as a driving force for net MC adhesion.
Collapse
Affiliation(s)
- Elena P. Moiseeva
- Institute for Lung Health, Dept. of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
- * E-mail:
| | - Kees R. Straatman
- Centre for Core Biotechnology Services, University of Leicester, Leicester, United Kingdom
| | - Mark L. Leyland
- Department of Biochemistry, University of Leicester, Leicester, United Kingdom
| | - Peter Bradding
- Institute for Lung Health, Dept. of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
40
|
Qiu Y, Luo X, Kan T, Zhang Y, Yu W, Wei Y, Shen N, Yi B, Jiang X. TGF-β upregulates miR-182 expression to promote gallbladder cancer metastasis by targeting CADM1. MOLECULAR BIOSYSTEMS 2014; 10:679-85. [PMID: 24445397 DOI: 10.1039/c3mb70479c] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
UNLABELLED Transforming growth factor β (TGF-β) plays important roles in tumor metastasis by regulating miRNAs expression. miR-182 is an important molecule in the regulation of cancer progression. The aim of the study is to assess the role of miR-182 in TGF-β-induced cancer metastasis. In the present study, we found that miR-182 levels are significantly upregulated in GBC tissues compared with normal controls, and miR-182 expression is remarkably increased in primary tumors that subsequently metastasized, when compared to those primary tumors that did not metastasize. TGF-β induces miR-182 expression in GBC cells, and overexpression of miR-182 promotes GBC cell migration and invasion, whereas miR-182 inhibition suppresses TGF-β-induced cancer cell migration and invasion. The blockage of miR-182 by a specific inhibitor effectively inhibits pulmonary metastases in vivo. We further identified that the cell adhesion molecule1 (CADM1) is a new target gene of miR-182. miR-182 negatively regulates CADM1 expression in vitro and in vivo. Importantly, re-expression of CADM1 in GBC cells partially abrogates miR-182-induced cell invasion. CONCLUSIONS miR-182 is an important mediator of GBC metastasis, thus offering a new target for the development of therapeutic agents against GBC.
Collapse
Affiliation(s)
- Yinghe Qiu
- The Second Department of Biliary Tract Surgery, Eastern Hepatobiliary Surgery Hospital, The Second Military Medicine University, 225 Changhai Road, Shanghai 200438, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Mimae T, Hagiyama M, Inoue T, Yoneshige A, Kato T, Okada M, Murakami Y, Ito A. Increased ectodomain shedding of lung epithelial cell adhesion molecule 1 as a cause of increased alveolar cell apoptosis in emphysema. Thorax 2013; 69:223-31. [PMID: 24092566 PMCID: PMC3933066 DOI: 10.1136/thoraxjnl-2013-203867] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Rationale Alveolar epithelial cell apoptosis and protease/antiprotease imbalance based proteolysis play central roles in the pathogenesis of pulmonary emphysema but molecular mechanisms underlying these two events are not yet clearly understood. Cell adhesion molecule 1 (CADM1) is a lung epithelial cell adhesion molecule in the immunoglobulin superfamily. It generates two membrane associated C terminal fragments (CTFs), αCTF and βCTF, through protease mediated ectodomain shedding. Objective To explore the hypothesis that more CADM1-CTFs are generated in emphysematous lungs through enhanced ectodomain shedding, and cause increased apoptosis of alveolar epithelial cells. Methods and results Western blot analyses revealed that CADM1-CTFs increased in human emphysematous lungs in association with increased ectodomain shedding. Increased apoptosis of alveolar epithelial cells in emphysematous lungs was confirmed by terminal nucleotide nick end labelling (TUNEL) assays. NCI-H441 lung epithelial cells expressing mature CADM1 but not CTFs were induced to express αCTF both endogenously (by shedding inducers phorbol ester and trypsin) and exogenously (by transfection). Cell fractionation, immunofluorescence, mitochondrial membrane potentiometric JC-1 dye labelling and TUNEL assays revealed that CADM1-αCTF was localised to mitochondria where it decreased mitochondrial membrane potential and increased cell apoptosis. A mutation in the intracytoplasmic domain abrogated all three abilities of αCTF. Conclusions CADM1 ectodomain shedding appeared to cause alveolar cell apoptosis in emphysematous lungs by producing αCTF that accumulated in mitochondria. These data link proteolysis to apoptosis, which are two landmark events in emphysema.
Collapse
Affiliation(s)
- Takahiro Mimae
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Graduate School of Biomedical Sciences, Hiroshima University, , Hiroshima, Japan
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Lei W, Liu HB, Wang SB, Zhou XM, Zheng SD, Guo KN, Ma BY, Xia YL, Tan WS, Liu XY, Wang YG. Tumor suppressor in lung cancer-1 (TSLC1) mediated by dual-regulated oncolytic adenovirus exerts specific antitumor actions in a mouse model. Acta Pharmacol Sin 2013; 34:531-40. [PMID: 23503473 DOI: 10.1038/aps.2012.196] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIM The tumor suppressor in lung cancer-1 (TSLC1) is a candidate tumor suppressor of lung cancer, and frequently inactivated in primary non-small cell lung cancer (NSCLC). In this study, we investigated the effects of TSLC1 mediated by a dual-regulated oncolytic adenovirus on lung cancer, and the mechanisms underlying the antitumor actions. METHODS The recombinant virus Ad·sp-E1A(Δ24)-TSLC1 was constructed by inserting the TSLC1 gene into the dual-regulated Ad·sp-E1A(Δ24) vector, which contained the survivin promoter and a 24 bp deletion within E1A. The antitumor effects of Ad·sp-E1A(Δ24)-TSLC1 were evaluated in NCI-H460, A549, and H1299 lung cancer cell lines and the normal fibroblast cell line MRC-5, as well as in A549 xenograft model in nude mice. Cell viability was assessed using MTT assay. The expression of TSLC1 and activation of the caspase signaling pathway were detected by Western blot analyses. The tumor tissues from the xenograft models were examined using H&E staining, IHC, TUNEL, and TEM analyses. RESULTS Infection of A549 lung cancer cells with Ad·sp-E1A(Δ24)-TSLC1 induced high level expression of TSLC1. Furthermore, the Ad·sp-E1A(Δ24)-TSLC1 virus dose-dependently suppressed the viability of NCI-H460, A549, and H1299 lung cancer cells, and did not affect MRC-5 normal fibroblast cells. Infection of NCI-H460, A549, and H1299 lung cancer cells with Ad·sp-E1A(Δ24)-TSLC1 induced apoptosis, and increased activation of caspase-8, caspase-3 and PARP. In A549 xenograft model in nude mice, intratumoral injection of Ad·sp-E1A(Δ24)-TSLC1 significantly suppressed the tumor volume, and increased the survival rate (from less than 15% to 87.5% at d 60). Histological studies showed that injection of Ad·sp-E1A(Δ24)-TSLC1 caused tumor cell apoptosis and virus particle propagation in tumor tissues. CONCLUSION The oncolytic adenovirus Ad·sp-E1A(Δ24)-TSLC1 exhibits specific antitumor effects, and is a promising agent for the treatment of lung cancer.
Collapse
|
43
|
Kikuchi S, Iwai M, Sakurai-Yageta M, Tsuboi Y, Ito T, Maruyama T, Tsuda H, Kanai Y, Onizuka M, Sato Y, Murakami Y. Expression of a splicing variant of the CADM1 specific to small cell lung cancer. Cancer Sci 2012; 103:1051-7. [PMID: 22429880 DOI: 10.1111/j.1349-7006.2012.02277.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Revised: 03/01/2012] [Accepted: 03/04/2012] [Indexed: 12/25/2022] Open
Abstract
CADM1, a member of the immunoglobulin superfamily cell adhesion molecule, acts as a tumor suppressor in a variety of human cancers. CADM1 is also ectopically expressed in adult T-cell leukemia (ATL), conferring an invasive phenotype characteristic to ATL. Therefore, CADM1 plays dual roles in human oncogenesis. Here, we investigate the roles of CADM1 in small cell lung cancer (SCLC). Immunohistochemistry demonstrates that 10 of 35 (29%) primary SCLC tumors express CADM1 protein. Western blotting and RT-PCR analyses reveal that CADM1 is significantly expressed in 11 of 14 SCLC cells growing in suspension cultures but in neither of 2 SCLC cells showing attached growth to plastic dishes, suggesting that CADM1 is involved in anchorage-independent growth in SCLC. In the present study, we demonstrate that SCLC expresses a unique splicing variant of CADM1 (variant 8/9) containing additional extracellular fragments corresponding to exon 9 in addition to variant 8, a common isoform in epithelia. Variant 8/9 of CADM1 is almost exclusively observed in SCLC and testis, although this variant protein localizes along the membrane and shows similar cell aggregation activity to variant 8. Interestingly, both variant 8/9 and variant 8 of CADM1 show enhanced tumorigenicity in nude mice when transfected into SBC5, a SCLC cell lacking CADM1. Inversely, suppression of CADM1 expression by shRNA reduced spheroid-like cell aggregation of NCI-H69, an SCLC cell expressing a high amount of CADM1. These findings suggest that CADM1 enhances the malignant features of SCLC, as is observed in ATL, and could provide a molecular marker specific to SCLC.
Collapse
Affiliation(s)
- Shinji Kikuchi
- Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Li QJ, Zhou L, Yang F, Wang GX, Zheng H, Wang DS, He Y, Dou KF. MicroRNA-10b promotes migration and invasion through CADM1 in human hepatocellular carcinoma cells. Tumour Biol 2012; 33:1455-65. [PMID: 22528944 DOI: 10.1007/s13277-012-0396-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Accepted: 03/30/2012] [Indexed: 12/15/2022] Open
Abstract
MicroRNA-10b (miR-10b) was recently reported to be dysregulated in some types of cancer and to play a role in invasion and metastasis. However, effects and potential mechanisms of action of miR-10b in the metastasis of hepatocellular carcinoma (HCC) have not been explored. In this study, we confirmed that miR-10b is highly expressed in metastatic HCC tissues and in metastatic HCC cell lines by qRT-PCR. Moreover, patients with higher miR-10b expression had significantly poorer overall survival, and high miR-10b expression was an independent predictor of poor prognosis. Inhibition of miR-10b reduced cell migration and invasion in MHCC97H cells, whereas over-expression of miR-10b in HepG2 cells increased cell migration and invasion. Bioinformatics and luciferase reporter assays revealed that miR-10b binds the 3'-UTR of CADM1 mRNA and represses its translation. Western blot and qRT-PCR showed that CADM1 is inhibited by miR-10b over-expression. Silencing of CADM1 resulted in substantially increased cell motility and invasion similar to that observed with over-expression of miR-10b in HepG2 cells. These results suggest that miR-10b may positively regulate the invasion and metastasis of HCC through targeting CADM1.
Collapse
Affiliation(s)
- Qing-jun Li
- Department of Hepatobiliary Surgery, Xijing Hospital, the Fourth Military Medical University, No. 17 Changle West Road, Xi'an, 710032, China
| | | | | | | | | | | | | | | |
Collapse
|
45
|
He G, Lei W, Wang S, Xiao R, Guo K, Xia Y, Zhou X, Zhang K, Liu X, Wang Y. Overexpression of tumor suppressor TSLC1 by a survivin-regulated oncolytic adenovirus significantly inhibits hepatocellular carcinoma growth. J Cancer Res Clin Oncol 2012; 138:657-70. [PMID: 22237452 DOI: 10.1007/s00432-011-1138-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 12/20/2011] [Indexed: 12/16/2022]
Abstract
PURPOSE Hepatocellular carcinoma (HCC) is one of the most common malignant tumors worldwide. Oncolytic viruses represent a promising therapeutic agent or vehicle to human cancers due to their ability of selectively lysing cancer cells but not in normal cells. TSLC1, a novel tumor suppressor gene, was loss in many human cancers including HCC, not in normal cells. The current study is focused on the antitumor effect of TSLC1-armed survivin-regulated oncolytic adenovirus for HCC and to explore their molecular mechanism. METHODS The expression of tumor suppressor TSLC1 and survivin was detected by quantitative PCR. The recombinant virus Ad.SP-E1A-E1B((Δ55))-TSLC1 (brief name as SD55-TSLC1) was constructed by inserting TSLC1 gene into the dual-regulated oncolytic adenovirus vector Ad.SP-E1A-E1B((Δ55)). Then, we performed the antitumor experiments of SD55-TSLC1 in vitro and in nude mice xenografted with Huh7 liver cancer. RESULTS The expression of TSLC1 was lower in HCC cells than in normal cells, which implied TSLC1 is a tumor suppressor of liver cancer. Survivin expression is higher in detected HCC cells than in normal cells. The SD55-TSLC1 exhibited an excellent antitumor effect on HCC cell growth in vitro but does no or little damage to normal liver cells. Animal experiment further confirmed that SD55-TSLC1 achieved significant inhibition of Huh7 liver cancer xenografted growth. Furthermore, the mechanism of antitumor efficacy by SD55-TSLC1 was elucidated to be due to the activation of caspase apoptotic pathway including the inducement of caspase-3, caspase-8, and poly (ADP-ribose) polymerase cleavage. This is the first report of TSLC1 by oncolytic adenovirus with an excellent antitumor effect to liver cancer growth. CONCLUSION These data suggest that an oncolytic adenovirus expressing TSLC1 is effective and support that SD55-TSLC1 may be a potent antitumoral agent for future clinical trials of liver cancer.
Collapse
MESH Headings
- Adenoviridae/genetics
- Adenoviridae/physiology
- Adenovirus E1A Proteins/genetics
- Animals
- Apoptosis/genetics
- Blotting, Western
- Caspases/metabolism
- Cell Adhesion Molecule-1
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Cell Line, Tumor
- Female
- Gene Expression Regulation, Neoplastic
- Genetic Vectors/genetics
- HEK293 Cells
- Host-Pathogen Interactions/genetics
- Humans
- Immunoglobulins/genetics
- Immunoglobulins/metabolism
- Inhibitor of Apoptosis Proteins/genetics
- Inhibitor of Apoptosis Proteins/metabolism
- Liver Neoplasms, Experimental/genetics
- Liver Neoplasms, Experimental/therapy
- Liver Neoplasms, Experimental/virology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Microscopy, Electron, Transmission
- Oncolytic Virotherapy/methods
- Oncolytic Viruses/genetics
- Oncolytic Viruses/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Survivin
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
- Viral Proteins/genetics
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Guoqing He
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, No. 2 Road Xiasha District, Hangzhou 310018, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Re-expression of cell adhesion molecule inhibits growth and induces apoptosis of human pancreatic cancer cell line PANC-1. ACTA ACUST UNITED AC 2011; 31:762-767. [PMID: 22173495 DOI: 10.1007/s11596-011-0673-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Indexed: 01/03/2023]
Abstract
This study examined the expression of cell adhesion molecule 1 (CADM1) in pancreatic cancer and the possible mechanism. The expression of CADM1 was detected by immunohistochemistry in tissues of pancreatic cancer, pancreatitis, and normal pancreas. The plasmid pcDNA3.1-Hygro(+)/CADM1 was transfected into PANC-1 cells (a pancreatic cancer cell line). The expression of CADM1 in the transfected cells was determined by RT-PCR and Western blotting. Cell growth was measured by the MTT method and cell apoptosis by flow cytometry. The results showed that CADM1 was weakly expressed in tissues of pancreatic cancer in contrast to its high expression in normal pancreatic and pancreatitis tissues. The expression level of CADM in pancreatic caner was intensely correlated with the differentiation degree, lymph node metastasis and TNM stages. The growth of CADM1-transfected PANC-1 cells was significantly suppressed in vitro by a G1 cell cycle arrest and apoptosis occurrence. It was concluded that re-expression of CADM1 inhibits the growth of pancreatic cancer cells and induces their apoptosis in vitro. As a tumor suppressor gene, CADM1 plays an important role in the occurrence, progression and metastasis of pancreatic cancer.
Collapse
|
47
|
Nawata H, Kashino G, Tano K, Daino K, Shimada Y, Kugoh H, Oshimura M, Watanabe M. Dysregulation of gene expression in the artificial human trisomy cells of chromosome 8 associated with transformed cell phenotypes. PLoS One 2011; 6:e25319. [PMID: 21980425 PMCID: PMC3183047 DOI: 10.1371/journal.pone.0025319] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 08/31/2011] [Indexed: 11/26/2022] Open
Abstract
A change in chromosome number, known as aneuploidy, is a common characteristic of cancer. Aneuploidy disrupts gene expression in human cancer cells and immortalized human epithelial cells, but not in normal human cells. However, the relationship between aneuploidy and cancer remains unclear. To study the effects of aneuploidy in normal human cells, we generated artificial cells of human primary fibroblast having three chromosome 8 (trisomy 8 cells) by using microcell-mediated chromosome transfer technique. In addition to decreased proliferation, the trisomy 8 cells lost contact inhibition and reproliferated after exhibiting senescence-like characteristics that are typical of transformed cells. Furthermore, the trisomy 8 cells exhibited chromosome instability, and the overall gene expression profile based on microarray analyses was significantly different from that of diploid human primary fibroblasts. Our data suggest that aneuploidy, even a single chromosome gain, can be introduced into normal human cells and causes, in some cases, a partial cancer phenotype due to a disruption in overall gene expression.
Collapse
Affiliation(s)
- Hisakatsu Nawata
- Laboratory of Radiation Biology, Research Reactor Institute, Kyoto University, Osaka, Japan
| | - Genro Kashino
- Advanced Molecular Imaging Center, Medical School, Oita University, Oita, Japan
| | - Keizo Tano
- Laboratory of Radiation Biology, Research Reactor Institute, Kyoto University, Osaka, Japan
| | - Kazuhiro Daino
- Experimental Radiobiology for Children's Health Research Group, Research Center for Radiation Protection, National Institute of Radiological Sciences, Chiba, Japan
| | - Yoshiya Shimada
- Experimental Radiobiology for Children's Health Research Group, Research Center for Radiation Protection, National Institute of Radiological Sciences, Chiba, Japan
| | - Hiroyuki Kugoh
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences, Tottori University, Tottori, Japan
| | - Mitsuo Oshimura
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences, Tottori University, Tottori, Japan
| | - Masami Watanabe
- Laboratory of Radiation Biology, Research Reactor Institute, Kyoto University, Osaka, Japan
- * E-mail:
| |
Collapse
|
48
|
Nagata M, Sakurai-Yageta M, Yamada D, Goto A, Ito A, Fukuhara H, Kume H, Morikawa T, Fukayama M, Homma Y, Murakami Y. Aberrations of a cell adhesion molecule CADM4 in renal clear cell carcinoma. Int J Cancer 2011; 130:1329-37. [DOI: 10.1002/ijc.26160] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 01/27/2011] [Indexed: 11/07/2022]
|
49
|
Dai Y, Wang J, Xia J, Hong Y, Chen N, Cheng B. Genome-wide transcriptional profiling analysis of all trans retinoic acid-treated tongue carcinoma SCC-9 cells. J Surg Oncol 2011; 104:830-5. [PMID: 21725974 DOI: 10.1002/jso.21998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 05/27/2011] [Indexed: 11/05/2022]
Abstract
BACKGROUND All trans retinoic acid (ATRA) is used as standard of care in promyelocytic leukemia. Not much is known about the gene expression profile in ATRA-treated tongue cancer cells. We performed a genome-wide transcriptional profiling of ATRA-treated tongue cancer cells to understand the pathways that mediate ATRA action in tongue cancer. METHODS We measured the effects of ATRA on the proliferation of SCC-9 human tongue carcinoma cells. The differential gene expression profile was measured by microarray analysis of untreated and ATRA-treated cells and expression of key genes was validated by real-time RT-PCR. RESULTS ATRA treatment (24 and 48 hr) significantly inhibited SCC-9 cell proliferation in a dose-dependent manner. SCC-9 cells treated for 48 hr with ATRA showed upregulation of 276 genes, including ANGPTL4, GDF15, ICAM1 and TUSC4, and downregulation of 43 genes, including CXCL10. Validation by real-time PCR showed a significant upregulation of intracellular adhesion molecule 1 (ICAM1) and downregulation of CXCL10 and IL32. CONCLUSIONS ATRA had an anti-tumor effect in tongue cancer cells. This effect is likely mediated via upregulation of ICAM1 and downregulation of CXCL10 and IL32.
Collapse
Affiliation(s)
- Yaohui Dai
- Department of Oral Medicine, Guanghua School of Stomatology and Institute of Stomatological Research, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | | | | | | | | | | |
Collapse
|
50
|
Aberrant expression of tumor suppressors CADM1 and 4.1B in invasive lesions of primary breast cancer. Breast Cancer 2011; 19:242-52. [PMID: 21526423 DOI: 10.1007/s12282-011-0272-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Accepted: 04/04/2011] [Indexed: 02/06/2023]
Abstract
BACKGROUND The tumor suppressor genes CADM1/TSLC1 and DAL-1/4.1B are frequently inactivated by promoter methylation in non-small cell lung cancer. The proteins they encode, CADM1 and 4.1B, form a complex in human epithelial cells and are involved in cell-cell adhesion. METHODS Expression of CADM1 and 4.1B proteins was examined by immunohistochemistry in 67 primary breast cancer and adjacent noncancerous tissues. CADM1 and 4.1B messenger RNA (mRNA) was detected by reverse-transcription polymerase chain reaction (RT-PCR). The methylation status of the CADM1 and 4.1B promoters was determined quantitatively by bisulfite treatment followed by pyrosequencing. RESULTS CADM1 and 4.1B protein signals were detected along the cell membrane in normal mammary epithelia. By contrast, 47 (70%) and 49 (73%) of 67 primary breast cancers showed aberrant CADM1 and 4.1B staining, respectively. Aberrant CADM1 staining was more frequently observed in pT2 and pT3 tumors and for stages II and III (P = 0.045 and P = 0.020, respectively), while aberrant 4.1B staining was more often observed in tumors with lymph node metastasis, for pT2 and pT3 tumors, and for stages II and III (P = 0.0058, P = 0.0098, and P = 0.0007, respectively). Furthermore, aberrant CADM1 and 4.1B expression was preferentially observed in invasive relative to noninvasive lesions from the same specimen (P = 0.036 and P = 0.0009, respectively). Finally, hypermethylation of CADM1 and 4.1B genes was detected in 46% and 42% of primary breast cancers, respectively. CONCLUSIONS Our findings suggest that aberrant CADM1 and 4.1B expression is involved in progression of breast cancer, especially in invasion into the stroma and metastasis.
Collapse
|