1
|
Coelho NM, Riahi P, Wang Y, Ali A, Norouzi M, Kotlyar M, Jurisica I, McCulloch CA. The major vault protein integrates adhesion-driven signals to regulate collagen remodeling. Cell Signal 2024; 124:111447. [PMID: 39368789 DOI: 10.1016/j.cellsig.2024.111447] [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: 08/22/2024] [Revised: 09/21/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
DDR1 interacts with fibrillar collagen and can affect β1 integrin-dependent signaling, but the mechanism that mediates functional interactions between these two different receptors is not defined. We searched for molecules that link DDR1 and β1 integrin-dependent signaling in response to collagen binding. The activation of DDR1 by binding to fibrillar collagen reduced by 5-fold, β1 integrin-dependent ERK phosphorylation that leads to MMP1 expression. In contrast, pharmacological inhibition of DDR1 or culturing cells on fibronectin restored ERK phosphorylation and MMP1 expression mediated by the β1 integrin. A phospho-site screen indicated that collagen-induced DDR1 activation inhibited β1 integrin-dependent ERK signaling by regulating autophosphorylation of focal adhesion kinase (FAK). Immunoprecipitation, mass spectrometry, and protein-protein interaction mapping showed that while DDR1 and FAK do not interact directly, the major vault protein (MVP) binds DDR1 and FAK depending on the substrate. MVP associated with DDR1 in cells expressing β1 integrin that were cultured on collagen. Knockdown of MVP restored ERK activation and MMP1 expression in DDR1-expressing cells cultured on collagen. Immunostaining of invasive cancers in human colon showed colocalization of DDR1 with MVP. These data indicate that MVP interactions with DDR1 and FAK contribute to the regulation of β1 integrin-dependent signaling pathways that drive collagen degradation.
Collapse
Affiliation(s)
- Nuno M Coelho
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Pardis Riahi
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Yongqiang Wang
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Aiman Ali
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Masoud Norouzi
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Max Kotlyar
- Osteoarthritis Research Program, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, UHN, Toronto, ON, Canada
| | - Igor Jurisica
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada; Osteoarthritis Research Program, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, UHN, Toronto, ON, Canada; Departments of Medical Biophysics and Computer Science, University of Toronto, ON, Canada
| | | |
Collapse
|
2
|
Wei Z, Li J, Zhong L, Yang D, Li W, Chen W, Zhou H, He Y, Song W, Wang B, Zeng L. DDR1 Drives Malignant Progression of Gastric Cancer by Suppressing HIF-1α Ubiquitination and Degradation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308395. [PMID: 39024501 PMCID: PMC11425230 DOI: 10.1002/advs.202308395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 06/07/2024] [Indexed: 07/20/2024]
Abstract
The extracellular matrix (ECM) has been demonstrated to be dysregulated and crucial for malignant progression in gastric cancer (GC), but the mechanism is not well understood. Here, that discoidin domain receptor 1 (DDR1), a principal ECM receptor, is recognized as a key driver of GC progression is reported. Mechanistically, DDR1 directly interacts with the PAS domain of hypoxia-inducible factor-1α (HIF-1α), suppresses its ubiquitination and subsequently strengthens its transcriptional regulation of angiogenesis. Additionally, DDR1 upregulation in GC cells promotes actin cytoskeleton reorganization by activating HIF-1α/ Ras Homolog Family Member A (RhoA)/Rho-associated protein kinase 1 (ROCK1) signaling, which in turn enhances the metastatic capacity. Pharmacological inhibition of DDR1 suppresses GC progression and angiogenesis in patient-derived xenograft (PDX) and organoid models. Taken together, this work first indicates the effects of the DDR1-HIF-1α axis on GC progression and reveals the related mechanisms, providing experimental evidence for DDR1 as a therapeutic target for GC.
Collapse
Affiliation(s)
- Zhewei Wei
- Department of Gastrointestinal SurgeryThe First Affiliated Hospital of Sun Yat‐sen University58 Zhongshan 2nd RoadGuangzhouGuangdong510080China
| | - Jin Li
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer ResearchScientific Research CenterThe BiobankThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityNo. 628 Zhenyuan RoadShenzhenGuangdong518107China
| | - Li Zhong
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer ResearchScientific Research CenterThe BiobankThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityNo. 628 Zhenyuan RoadShenzhenGuangdong518107China
| | - Dongjie Yang
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer ResearchScientific Research CenterThe BiobankThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityNo. 628 Zhenyuan RoadShenzhenGuangdong518107China
| | - Wuguo Li
- Laboratory Animal CenterThe First Affiliated HospitalSun Yat‐sen University58 Zhongshan 2nd RoadGuangzhou510080China
| | - Wei Chen
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer ResearchScientific Research CenterThe BiobankThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityNo. 628 Zhenyuan RoadShenzhenGuangdong518107China
| | - Hao Zhou
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer ResearchScientific Research CenterThe BiobankThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityNo. 628 Zhenyuan RoadShenzhenGuangdong518107China
| | - Yulong He
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer ResearchScientific Research CenterThe BiobankThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityNo. 628 Zhenyuan RoadShenzhenGuangdong518107China
| | - Wu Song
- Department of Gastrointestinal SurgeryThe First Affiliated Hospital of Sun Yat‐sen University58 Zhongshan 2nd RoadGuangzhouGuangdong510080China
| | - Boyan Wang
- Reproductive Medicine CenterThe First Affiliated Hospital of Sun Yat‐sen University58 Zhongshan 2nd RoadGuangzhouGuangdong510080China
| | - Leli Zeng
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer ResearchScientific Research CenterThe BiobankThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityNo. 628 Zhenyuan RoadShenzhenGuangdong518107China
| |
Collapse
|
3
|
Sawant M, Wang F, Koester J, Niehoff A, Nava MM, Lundgren-Akerlund E, Gullberg D, Leitinger B, Wickström S, Eckes B, Krieg T. Ablation of integrin-mediated cell-collagen communication alleviates fibrosis. Ann Rheum Dis 2023; 82:1474-1486. [PMID: 37479494 DOI: 10.1136/ard-2023-224129] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/06/2023] [Indexed: 07/23/2023]
Abstract
OBJECTIVES Activation of fibroblasts is a hallmark of fibrotic processes. Besides cytokines and growth factors, fibroblasts are regulated by the extracellular matrix environment through receptors such as integrins, which transduce biochemical and mechanical signals enabling cells to mount appropriate responses according to biological demands. The aim of this work was to investigate the in vivo role of collagen-fibroblast interactions for regulating fibroblast functions and fibrosis. METHODS Triple knockout (tKO) mice with a combined ablation of integrins α1β1, α2β1 and α11β1 were created to address the significance of integrin-mediated cell-collagen communication. Properties of primary dermal fibroblasts lacking collagen-binding integrins were delineated in vitro. Response of the tKO mice skin to bleomycin induced fibrotic challenge was assessed. RESULTS Triple integrin-deficient mice develop normally, are transiently smaller and reveal mild alterations in mechanoresilience of the skin. Fibroblasts from these mice in culture show defects in cytoskeletal architecture, traction stress generation, matrix production and organisation. Ablation of the three integrins leads to increased levels of discoidin domain receptor 2, an alternative receptor recognising collagens in vivo and in vitro. However, this overexpression fails to compensate adhesion and spreading defects on collagen substrates in vitro. Mice lacking collagen-binding integrins show a severely attenuated fibrotic response with impaired mechanotransduction, reduced collagen production and matrix organisation. CONCLUSIONS The data provide evidence for a crucial role of collagen-binding integrins in fibroblast force generation and differentiation in vitro and for matrix deposition and tissue remodelling in vivo. Targeting fibroblast-collagen interactions might represent a promising therapeutic approach to regulate connective tissue deposition in fibrotic diseases.
Collapse
Affiliation(s)
- Mugdha Sawant
- Translational Matrix Biology, University of Cologne, Cologne, Germany
| | - Fang Wang
- Translational Matrix Biology, University of Cologne, Cologne, Germany
| | - Janis Koester
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Anja Niehoff
- Institute of Biomechanics and Orthopaedics, German Sport University, Cologne, Germany
- Cologne Center for Musculoskeletal Biomechanics (CCMB), University of Cologne, Medical Faculty, Cologne, Germany
| | - Michele M Nava
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | | | | | - Sara Wickström
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Helsinki Institute of Life Science, Biomedicum Helsinki, Helsinki, Finland
| | - Beate Eckes
- Translational Matrix Biology, University of Cologne, Cologne, Germany
| | - Thomas Krieg
- Translational Matrix Biology, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|
4
|
Li X, Chen H, Zhang D. Discoidin domain receptor 1 may be involved in biological barrier homeostasis. J Clin Pharm Ther 2022; 47:2397-2407. [PMID: 35665520 DOI: 10.1111/jcpt.13705] [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: 12/04/2021] [Revised: 04/08/2022] [Accepted: 04/25/2022] [Indexed: 12/24/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Discoidin domain receptor 1 (DDR1) is a receptor tyrosine kinase involved in the pathological processes of several diseases, such as keloid formation, renal fibrosis, atherosclerosis, tumours, and inflammatory processes. The biological barrier is the first line of defence against pathogens, and its disruption is closely related to diseases. In this review, we attempt to elucidate the relationship between DDR1 and the biological barrier, explore the potential biological value of DDR1, and review the current research status and clinical potential of DDR1-selective inhibitors. METHODS We conducted an extensive literature search on PubMed to collect studies on the relevance of DDR1 to biological barriers and DDR1-selective inhibitors. With these studies, we explored the relationship between DDR1 and biological barriers and briefly reviewed representative DDR1-selective inhibitors that have been reported in recent years. RESULTS AND DISCUSSION First, the review of the potential mechanisms by which DDR1 regulates biological barriers, including the epithelial, vascular, glomerular filtration, blood-labyrinth, and blood-brain barriers. In the body, DDR1 dysfunction and aberrant expression may be involved in the homeostasis of the biological barrier. Secondly, the review of DDR1 inhibitors reported in recent years shows that DDR1-targeted inhibition is an attractive and promising pharmacological intervention. WHAT IS NEW AND CONCLUSIONS This review shows that DDR1 is involved in various physiological and pathological processes and in the regulation of biological barrier homeostasis. However, studies on DDR1 and biological barriers are still scarce, and further studies are needed to elucidate their specific mechanisms. The development of targeted inhibitors provides a new direction and idea to study the mechanism of DDR1.
Collapse
Affiliation(s)
- Xiaoli Li
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Huiling Chen
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Dekui Zhang
- Department of Gastroenterology, Key Laboratory of Digestive Diseases, LanZhou University Second Hospital, LanZhou University, Lanzhou, China
| |
Collapse
|
5
|
Madison J, Wilhelm K, Meehan DT, Delimont D, Samuelson G, Cosgrove D. Glomerular basement membrane deposition of collagen α1(III) in Alport glomeruli by mesangial filopodia injures podocytes via aberrant signaling through DDR1 and integrin α2β1. J Pathol 2022; 258:26-37. [PMID: 35607980 PMCID: PMC9378723 DOI: 10.1002/path.5969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/29/2022] [Accepted: 05/20/2022] [Indexed: 11/20/2022]
Abstract
In Alport mice, activation of the endothelin A receptor (ETA R) in mesangial cells results in sub-endothelial invasion of glomerular capillaries by mesangial filopodia. Filopodia deposit mesangial matrix in the glomerular basement membrane (GBM), including laminin 211 which activates NF-κB, resulting in induction of inflammatory cytokines. Herein we show that collagen α1(III) is also deposited in the GBM. Collagen α1(III) localized to the mesangium in wild-type mice and was found in both the mesangium and the GBM in Alport mice. We show that collagen α1(III) activates discoidin domain receptor family, member 1 (DDR1) receptors both in vitro and in vivo. To elucidate whether collagen α1(III) might cause podocyte injury, cultured murine Alport podocytes were overlaid with recombinant collagen α1(III), or not, for 24 h and RNA was analyzed by RNA sequencing (RNA-seq). These same cells were subjected to siRNA knockdown for integrin α2 or DDR1 and the RNA was analyzed by RNA-seq. Results were validated in vivo using RNA-seq from RNA isolated from wild-type and Alport mouse glomeruli. Numerous genes associated with podocyte injury were up- or down-regulated in both Alport glomeruli and cultured podocytes treated with collagen α1(III), 18 of which have been associated previously with podocyte injury or glomerulonephritis. The data indicate α2β1 integrin/DDR1 co-receptor signaling as the dominant regulatory mechanism. This may explain earlier studies where deletion of either DDR1 or α2β1 integrin in Alport mice ameliorates renal pathology. © 2022 Boys Town National Research Hospital. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
|
6
|
Borza CM, Bolas G, Zhang X, Browning Monroe MB, Zhang MZ, Meiler J, Skwark MJ, Harris RC, Lapierre LA, Goldenring JR, Hook M, Rivera J, Brown KL, Leitinger B, Tyska MJ, Moser M, Böttcher RT, Zent R, Pozzi A. The Collagen Receptor Discoidin Domain Receptor 1b Enhances Integrin β1-Mediated Cell Migration by Interacting With Talin and Promoting Rac1 Activation. Front Cell Dev Biol 2022; 10:836797. [PMID: 35309920 PMCID: PMC8928223 DOI: 10.3389/fcell.2022.836797] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/04/2022] [Indexed: 01/17/2023] Open
Abstract
Integrins and discoidin domain receptors (DDRs) 1 and 2 promote cell adhesion and migration on both fibrillar and non fibrillar collagens. Collagen I contains DDR and integrin selective binding motifs; however, the relative contribution of these two receptors in regulating cell migration is unclear. DDR1 has five isoforms (DDR1a-e), with most cells expressing the DDR1a and DDR1b isoforms. We show that human embryonic kidney 293 cells expressing DDR1b migrate more than DDR1a expressing cells on DDR selective substrata as well as on collagen I in vitro. In addition, DDR1b expressing cells show increased lung colonization after tail vein injection in nude mice. DDR1a and DDR1b differ from each other by an extra 37 amino acids in the DDR1b cytoplasmic domain. Interestingly, these 37 amino acids contain an NPxY motif which is a central control module within the cytoplasmic domain of β integrins and acts by binding scaffold proteins, including talin. Using purified recombinant DDR1 cytoplasmic tail proteins, we show that DDR1b directly binds talin with higher affinity than DDR1a. In cells, DDR1b, but not DDR1a, colocalizes with talin and integrin β1 to focal adhesions and enhances integrin β1-mediated cell migration. Moreover, we show that DDR1b promotes cell migration by enhancing Rac1 activation. Mechanistically DDR1b interacts with the GTPase-activating protein (GAP) Breakpoint cluster region protein (BCR) thus reducing its GAP activity and enhancing Rac activation. Our study identifies DDR1b as a major driver of cell migration and talin and BCR as key players in the interplay between integrins and DDR1b in regulating cell migration.
Collapse
Affiliation(s)
- Corina M. Borza
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN, United States
| | - Gema Bolas
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN, United States
| | - Xiuqi Zhang
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN, United States
| | | | - Ming-Zhi Zhang
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN, United States
| | - Jens Meiler
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
- Leipzig University Medical School, Institute for Drug Discovery, Leipzig, Germany
| | - Marcin J. Skwark
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - Raymond C. Harris
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN, United States
| | - Lynne A. Lapierre
- Department of Surgery, Vanderbilt University, Nashville, TN, United States
- Veterans Affairs Hospital, Nashville, TN, United States
| | - James R. Goldenring
- Department of Surgery, Vanderbilt University, Nashville, TN, United States
- Veterans Affairs Hospital, Nashville, TN, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
| | - Magnus Hook
- Texas A&M Health Science Center Institute of Biosciences and Technology, Houston, TX, United States
| | - Jose Rivera
- Texas A&M Health Science Center Institute of Biosciences and Technology, Houston, TX, United States
| | - Kyle L. Brown
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN, United States
| | - Birgit Leitinger
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Matthew J. Tyska
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
| | - Markus Moser
- Department for Molecular Medicine, Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | - Ralph T. Böttcher
- Department for Molecular Medicine, Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | - Roy Zent
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN, United States
- Veterans Affairs Hospital, Nashville, TN, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
| | - Ambra Pozzi
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN, United States
- Veterans Affairs Hospital, Nashville, TN, United States
| |
Collapse
|
7
|
Chen L, Kong X, Fang Y, Paunikar S, Wang X, Brown JAL, Bourke E, Li X, Wang J. Recent Advances in the Role of Discoidin Domain Receptor Tyrosine Kinase 1 and Discoidin Domain Receptor Tyrosine Kinase 2 in Breast and Ovarian Cancer. Front Cell Dev Biol 2021; 9:747314. [PMID: 34805157 PMCID: PMC8595330 DOI: 10.3389/fcell.2021.747314] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Discoidin domain receptor tyrosine kinases (DDRs) are a class of receptor tyrosine kinases (RTKs), and their dysregulation is associated with multiple diseases (including cancer, chronic inflammatory conditions, and fibrosis). The DDR family members (DDR1a-e and DDR2) are widely expressed, with predominant expression of DDR1 in epithelial cells and DDR2 in mesenchymal cells. Structurally, DDRs consist of three regions (an extracellular ligand binding domain, a transmembrane domain, and an intracellular region containing a kinase domain), with their kinase activity induced by receptor-specific ligand binding. Collagen binding to DDRs stimulates DDR phosphorylation activating kinase activity, signaling to MAPK, integrin, TGF-β, insulin receptor, and Notch signaling pathways. Abnormal DDR expression is detected in a range of solid tumors (including breast, ovarian, cervical liver, gastric, colorectal, lung, and brain). During tumorigenesis, abnormal activation of DDRs leads to invasion and metastasis, via dysregulation of cell adhesion, migration, proliferation, secretion of cytokines, and extracellular matrix remodeling. Differential expression or mutation of DDRs correlates with pathological classification, clinical characteristics, treatment response, and prognosis. Here, we discuss the discovery, structural characteristics, organizational distribution, and DDR-dependent signaling. Importantly, we highlight the key role of DDRs in the development and progression of breast and ovarian cancer.
Collapse
Affiliation(s)
- Li Chen
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Breast Surgical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Shishir Paunikar
- Discipline of Pathology, School of Medicine, Lambe Institute for Translational Research, National University of Ireland Galway, Galway, Ireland
| | - Xiangyu Wang
- Department of Breast Surgical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - James A. L. Brown
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
- Health Research Institute, University of Limerick, Limerick, Ireland
| | - Emer Bourke
- Discipline of Pathology, School of Medicine, Lambe Institute for Translational Research, National University of Ireland Galway, Galway, Ireland
| | - Xingrui Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Massimini M, Romanucci M, De Maria R, Della Salda L. An Update on Molecular Pathways Regulating Vasculogenic Mimicry in Human Osteosarcoma and Their Role in Canine Oncology. Front Vet Sci 2021; 8:722432. [PMID: 34631854 PMCID: PMC8494780 DOI: 10.3389/fvets.2021.722432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/23/2021] [Indexed: 01/16/2023] Open
Abstract
Canine tumors are valuable comparative models for human counterparts, especially to explore novel biomarkers and to understand pathways and processes involved in metastasis. Vasculogenic mimicry (VM) is a unique property of malignant cancer cells which promote metastasis. Thus, it represents an opportunity to investigate both the molecular mechanisms and the therapeutic targets of a crucial phenotypic malignant switch. Although this biological process has been largely investigated in different human cancer types, including osteosarcoma, it is still largely unknown in veterinary pathology, where it has been mainly explored in canine mammary tumors. The presence of VM in human osteosarcoma is associated with poor clinical outcome, reduced patient survival, and increased risk of metastasis and it shares the main pathways involved in other type of human tumors. This review illustrates the main findings concerning the VM process in human osteosarcoma, search for the related current knowledge in canine pathology and oncology, and potential involvement of multiple pathways in VM formation, in order to provide a basis for future investigations on VM in canine tumors.
Collapse
|
9
|
Feng Y, Cai H, Huang X, Li Z, Chi Z, Ge RL. Active MT1-MMP is tethered to collagen fibers in DDR2-containing remnants. Gene 2021; 788:145673. [PMID: 33882324 DOI: 10.1016/j.gene.2021.145673] [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: 05/07/2020] [Revised: 08/17/2020] [Accepted: 04/15/2021] [Indexed: 11/25/2022]
Abstract
Type I collagen is a major extracellular matrix (ECM) component in the interstitial stroma of solid tumors, and it represents the first barrier against tumor cell invasion after basement-membrane degradation. The collagen receptors that convey molecular signals into the cells are collagen-binding discoidin domain receptors (DDRs) and integrins. Collagen-activated DDR2 clusters form DDR2-containing remnants in an integrin-dependent manner in three-dimensional (3D) collagen matrix. Although DDR2-containing remnants in the collagen matrix may generate sustained perturbation to ECM remodeling, the molecular components and function of the remnants are largely unknown. Here we determined the interaction and co-localization between DDR2 and membrane type I-matrix metalloproteinase (MT1-MMP) in the cells and the DDR2-containing remnants on collagen fibers, and we found that MT1-MMP was co-tethered to collagen fibers in the remnants. These collagen fiber-associated MT1-MMP remained active. Furthermore, DDR2 enhanced MT1-MMP proteolytic activity. These results demonstrate that DDR2 ensures the remnant-associated MT1-MMP to continue the degradation of ECM in addition to pericellular ECM degradation mediated by cell surface tethered MT1-MMP. Thus, our findings reveal a new alternative ECM degradation mechanism mediated by MT1-MMP in the DDR2-containing remnants.
Collapse
Affiliation(s)
- Yunfeng Feng
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai 810001, China; Innate Gene Inc. Beijing 100085, China.
| | - Hao Cai
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai 810001, China; The Fifth Hospital of Qinghai Province, Xining, Qinghai 810001, China
| | | | | | | | - Ri-Li Ge
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai 810001, China.
| |
Collapse
|
10
|
DDR1 and DDR2: a review on signaling pathway and small molecule inhibitors as an anticancer agent. Med Chem Res 2021. [DOI: 10.1007/s00044-020-02694-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
11
|
Zangouei AS, Barjasteh AH, Rahimi HR, Mojarrad M, Moghbeli M. Role of tyrosine kinases in bladder cancer progression: an overview. Cell Commun Signal 2020; 18:127. [PMID: 32795296 PMCID: PMC7427778 DOI: 10.1186/s12964-020-00625-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/02/2020] [Indexed: 12/20/2022] Open
Abstract
Background Bladder cancer (BCa) is a frequent urothelial malignancy with a high ratio of morbidity and mortality. Various genetic and environmental factors are involved in BCa progression. Since, majority of BCa cases are diagnosed after macroscopic clinical symptoms, it is required to find efficient markers for the early detection. Receptor tyrosine-kinases (RTKs) and non-receptor tyrosine-kinases (nRTKs) have pivotal roles in various cellular processes such as growth, migration, differentiation, and metabolism through different signaling pathways. Tyrosine-kinase deregulations are observed during tumor progressions via mutations, amplification, and chromosomal abnormalities which introduces these factors as important candidates of anti-cancer therapies. Main body For the first time in present review we have summarized all of the reported tyrosine-kinases which have been significantly associated with the clinicopathological features of BCa patients. Conclusions This review highlights the importance of tyrosine-kinases as critical markers in early detection and therapeutic purposes among BCa patients and clarifies the molecular biology of tyrosine-kinases during BCa progression and metastasis. Video abstract
Collapse
Affiliation(s)
- Amir Sadra Zangouei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Hossein Barjasteh
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Rahimi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Mojarrad
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
12
|
Lorusso G, Rüegg C, Kuonen F. Targeting the Extra-Cellular Matrix-Tumor Cell Crosstalk for Anti-Cancer Therapy: Emerging Alternatives to Integrin Inhibitors. Front Oncol 2020; 10:1231. [PMID: 32793493 PMCID: PMC7387567 DOI: 10.3389/fonc.2020.01231] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/16/2020] [Indexed: 12/11/2022] Open
Abstract
The extracellular matrix (ECM) is a complex network composed of a multitude of different macromolecules. ECM components typically provide a supportive structure to the tissue and engender positional information and crosstalk with neighboring cells in a dynamic reciprocal manner, thereby regulating tissue development and homeostasis. During tumor progression, tumor cells commonly modify and hijack the surrounding ECM to sustain anchorage-dependent growth and survival, guide migration, store pro-tumorigenic cell-derived molecules and present them to enhance receptor activation. Thereby, ECM potentially supports tumor progression at various steps from initiation, to local growth, invasion, and systemic dissemination and ECM-tumor cells interactions have long been considered promising targets for cancer therapy. Integrins represent key surface receptors for the tumor cell to sense and interact with the ECM. Yet, attempts to therapeutically impinge on these interactions using integrin inhibitors have failed to deliver anticipated results, and integrin inhibitors are still missing in the emerging arsenal of drugs for targeted therapies. This paradox situation should urge the field to reconsider the role of integrins in cancer and their targeting, but also to envisage alternative strategies. Here, we review the therapeutic targets implicated in tumor cell adhesion to the ECM, whose inhibitors are currently in clinical trials and may offer alternatives to integrin inhibition.
Collapse
Affiliation(s)
- Girieca Lorusso
- Experimental and Translational Oncology, Department of Oncology Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Curzio Rüegg
- Experimental and Translational Oncology, Department of Oncology Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - François Kuonen
- Department of Dermatology and Venereology, Hôpital de Beaumont, Lausanne University Hospital Center, Lausanne, Switzerland
| |
Collapse
|
13
|
Yeh YC, Lin HH, Tang MJ. Dichotomy of the function of DDR1 in cells and disease progression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:118473. [PMID: 30954568 DOI: 10.1016/j.bbamcr.2019.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 12/18/2022]
Abstract
Discoidin domain receptors DDR1 and DDR2 are collagen receptor tyrosine kinases that have many roles in tissue development and disease progression. Under physiological conditions, DDR1 is predominantly expressed in epithelial cells and functions to maintain cell differentiation and tissue homeostasis. A switch in expression from DDR1 to DDR2 occurs during epithelial-to-mesenchymal transition. However, opposite effects of DDR1 are reported to be involved in the progression of cancer and fibrotic diseases. Accumulating evidence suggests that DDR1 is involved in pro-metastasis and pro-survival signals. This review summarizes the roles of DDR1 in epithelial cell differentiation, cell migration, cancer progression and tissues fibrosis and highlights how the dichotomous functions of DDR1 may relevant to different cell types and statues. Elucidation of the underlying mechanism of the dichotomous functions of DDR1 will help to develop DDR1 as a therapeutic target.
Collapse
Affiliation(s)
- Yi-Chun Yeh
- International Center for Wound Repair and Regeneration, Tainan, Taiwan
| | - Hsi-Hui Lin
- International Center for Wound Repair and Regeneration, Tainan, Taiwan; Department of Physiology, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Jer Tang
- International Center for Wound Repair and Regeneration, Tainan, Taiwan; Department of Physiology, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
14
|
Søgaard PP, Ito N, Sato N, Fujita Y, Matter K, Itoh Y. Epithelial polarization in 3D matrix requires DDR1 signaling to regulate actomyosin contractility. Life Sci Alliance 2019; 2:2/1/e201800276. [PMID: 30760555 PMCID: PMC6374992 DOI: 10.26508/lsa.201800276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/07/2019] [Accepted: 02/08/2019] [Indexed: 01/19/2023] Open
Abstract
For epithelial cells to establish epithelial polarity in a 3D matrix, signaling of a collagen receptor tyrosine kinase, DDR1, plays a crucial role. DDR1 signaling controls actomyosin contractility at the cell–cell junction through suppression of ROCK activity. Epithelial cells form sheets and tubules in various epithelial organs and establish apicobasal polarity and asymmetric vesicle transport to provide functionality in these structures. However, the molecular mechanisms that allow epithelial cells to establish polarity are not clearly understood. Here, we present evidence that the kinase activity of the receptor tyrosine kinase for collagen, discoidin domain receptor 1 (DDR1), is required for efficient establishment of epithelial polarity, proper asymmetric protein secretion, and execution of morphogenic programs. Lack of DDR1 protein or inhibition of DDR1 kinase activity disturbed tubulogenesis, cystogenesis, and the establishment of epithelial polarity and caused defects in the polarized localization of membrane-type 1 matrix metalloproteinase (MT1-MMP), GP135, primary cilia, laminin, and the Golgi apparatus. Disturbed epithelial polarity and cystogenesis upon DDR1 inhibition was caused by excess ROCK (rho-associated, coiled-coil-containing protein kinase)-driven actomyosin contractility, and pharmacological inhibition of ROCK was sufficient to correct these defects. Our data indicate that a DDR1-ROCK signaling axis is essential for the efficient establishment of epithelial polarity.
Collapse
Affiliation(s)
| | - Noriko Ito
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Nanami Sato
- Institute for Genetic Medicine, Division of Molecular Oncology, Hokkaido University, Sapporo, Japan
| | - Yasuyuki Fujita
- Institute for Genetic Medicine, Division of Molecular Oncology, Hokkaido University, Sapporo, Japan
| | - Karl Matter
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Yoshifumi Itoh
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
15
|
Li T, Liu J, Cai H, Wang B, Feng Y, Liu J. Incorporation of DDR2 clusters into collagen matrix via integrin-dependent posterior remnant tethering. Int J Biol Sci 2018; 14:654-666. [PMID: 29904280 PMCID: PMC6001655 DOI: 10.7150/ijbs.24765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022] Open
Abstract
Cell-matrix interactions play critical roles in cell adhesion, tissue remodeling and cancer metastasis. Discoidin domain receptor 2 (DDR2) is a collagen receptor belonging to receptor tyrosine kinase (RTK) family. It is a powerful regulator of collagen deposition in the extracellular matrix (ECM). Although the oligomerization of DDR extracellular domain (ECD) proteins can affect matrix remodeling by inhibiting fibrillogenesis, it is still unknown how cellular DDR2 is incorporated into collagen matrix. Using 3-dimentional (3D) imaging for migrating cells, we identified a novel mechanism that explains how DDR2 incorporating into collagen matrix, which we named as posterior remnant tethering. We followed the de novo formation of these remnants and identified that DDR2 clusters formed at the retracting phase of a pseudopodium, then these clusters were tethered to fibrillar collagen and peeled off from the cell body to generate DDR2 containing posterior remnants. Inhibition of β1-integrin or Rac1 activity abrogated the remnant formation. Thus, our findings unveil a special cellular mechanism for DDR2 clusters incorporating into collagen matrix in an integrin-dependent manner.
Collapse
Affiliation(s)
- Tingting Li
- Jiangsu key lab of Drug Screening, Jiangsu key lab of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing 210009, China
| | - Jin'e Liu
- Jiangsu key lab of Drug Screening, Jiangsu key lab of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing 210009, China
| | - Hao Cai
- Research Center for High Altitude Medicine, Qing Hai University, Xining 810001, China
| | - Baomei Wang
- Institute of Virology, Wenzhou University, Wenzhou, 325000, China
| | | | - Jun Liu
- Jiangsu key lab of Drug Screening, Jiangsu key lab of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
16
|
El Azreq MA, Kadiri M, Boisvert M, Pagé N, Tessier PA, Aoudjit F. Discoidin domain receptor 1 promotes Th17 cell migration by activating the RhoA/ROCK/MAPK/ERK signaling pathway. Oncotarget 2018; 7:44975-44990. [PMID: 27391444 PMCID: PMC5216699 DOI: 10.18632/oncotarget.10455] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 06/13/2016] [Indexed: 12/20/2022] Open
Abstract
Effector T cell migration through the tissue extracellular matrix (ECM) is an important step of the adaptive immune response and in the development of inflammatory diseases. However, the mechanisms involved in this process are still poorly understood. In this study, we addressed the role of a collagen receptor, the discoidin domain receptor 1 (DDR1), in the migration of Th17 cells. We showed that the vast majority of human Th17 cells express DDR1 and that silencing DDR1 or using the blocking recombinant receptor DDR1:Fc significantly reduced their motility and invasion in three-dimensional (3D) collagen. DDR1 promoted Th17 migration by activating RhoA/ROCK and MAPK/ERK signaling pathways. Interestingly, the RhoA/ROCK signaling module was required for MAPK/ERK activation. Finally, we showed that DDR1 is important for the recruitment of Th17 cells into the mouse dorsal air pouch containing the chemoattractant CCL20. Collectively, our results indicate that DDR1, via the activation of RhoA/ROCK/MAPK/ERK signaling axis, is a key pathway of effector T cell migration through collagen of perivascular tissues. As such, DDR1 can contribute to the development of Th17-dependent inflammatory diseases.
Collapse
Affiliation(s)
- Mohammed-Amine El Azreq
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Québec, QC, Canada
| | - Maleck Kadiri
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Québec, QC, Canada
| | - Marc Boisvert
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Québec, QC, Canada
| | - Nathalie Pagé
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Québec, QC, Canada
| | - Philippe A Tessier
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Québec, QC, Canada.,Département de Microbiologie-Immunologie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Fawzi Aoudjit
- Axe de Recherche sur les Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Québec, QC, Canada.,Département de Microbiologie-Immunologie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| |
Collapse
|
17
|
Hur H, Ham IH, Lee D, Jin H, Aguilera KY, Oh HJ, Han SU, Kwon JE, Kim YB, Ding K, Brekken RA. Discoidin domain receptor 1 activity drives an aggressive phenotype in gastric carcinoma. BMC Cancer 2017; 17:87. [PMID: 28143619 PMCID: PMC5286810 DOI: 10.1186/s12885-017-3051-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 01/10/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Discoidin domain receptor 1 (DDR1), a receptor tyrosine kinase that utilizes collagen as a ligand, is a key molecule in the progression of solid tumors as it regulates the interaction of cancer cells with the tumor stroma. However, the clinical relevance of DDR1 expression in gastric carcinoma is yet to be investigated. Here, we assessed the role of DDR1 in mediating the aggressive phenotype of gastric carcinoma and its potential as a therapeutic target. METHODS We conducted DDR1 immunohistochemistry using a tissue microarray of 202 gastric carcinoma specimens. We examined the effect of collagen-induced activation of DDR1 on cell signaling, tumorigenesis, and cell migration in gastric cancer cell lines, and tumor growth in a xenograft animal model of gastric cancer. RESULTS Our results showed that 50.5% of gastric cancer tissues are positive for DDR1 expression, and positive DDR1 expression was significantly correlated with a poor prognosis (P = 0.015). In a subgroup analysis, DDR1 expression was prognostically meaningful only in patients receiving adjuvant treatment (P = 0.013). We also demonstrated that collagen was able to activate DDR1 and increase the clonogenicity and migration of gastric cancer cells. We observed that a DDR1 inhibitor, 7rh benzamide, suppressed tumor growth in gastric cancer xenografts. CONCLUSIONS Our findings suggest a key role for DDR1 signaling in mediating the aggressive phenotype of gastric carcinoma. Importantly, inhibition of DDR1 is an attractive strategy for gastric carcinoma therapy.
Collapse
Affiliation(s)
- Hoon Hur
- Division of Gastrointestinal Surgery, Department of Surgery, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, 443-380, South Korea. .,Brain Korea 21 Plus Research Center for Biomedical Sciences, Ajou University, Suwon, Korea. .,Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX, 75390-8593, USA.
| | - In-Hye Ham
- Division of Gastrointestinal Surgery, Department of Surgery, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, 443-380, South Korea.,Brain Korea 21 Plus Research Center for Biomedical Sciences, Ajou University, Suwon, Korea
| | - Dakeun Lee
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | - Hyejin Jin
- Division of Gastrointestinal Surgery, Department of Surgery, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, 443-380, South Korea.,Brain Korea 21 Plus Research Center for Biomedical Sciences, Ajou University, Suwon, Korea
| | - Kristina Y Aguilera
- Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX, 75390-8593, USA
| | - Hye Jeong Oh
- Division of Gastrointestinal Surgery, Department of Surgery, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, 443-380, South Korea
| | - Sang-Uk Han
- Division of Gastrointestinal Surgery, Department of Surgery, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, 443-380, South Korea
| | - Ji Eun Kwon
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | - Young-Bae Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | - Ke Ding
- State Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Rolf A Brekken
- Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX, 75390-8593, USA.
| |
Collapse
|
18
|
DDR1 promotes E-cadherin stability via inhibition of integrin-β1-Src activation-mediated E-cadherin endocytosis. Sci Rep 2016; 6:36336. [PMID: 27824116 PMCID: PMC5099905 DOI: 10.1038/srep36336] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 10/14/2016] [Indexed: 01/08/2023] Open
Abstract
Discoidin domain receptor 1 (DDR1), a receptor tyrosine kinase of collagen, is primarily expressed in epithelial cells. Activation of DDR1 stabilises E-cadherin located on the cell membrane; however, the detailed mechanism of DDR1-stabilised E-cadherin remains unclear. We performed DDR1 knockdown (Sh-DDR1) on Mardin-Darby canine kidney cells to investigate the mechanism of DDR1-stabilised E-cadherin. Sh-DDR1 decreased junctional localisation, increased endocytosis of E-cadherin, and increased physical interactions between E-cadherin and clathrin. Treatment of the dynamin inhibitor Dyngo 4a suppressed Sh-DDR1-induced E-cadherin endocytosis. In addition, the phosphorylation level of Src tyrosine 418 was increased in Sh-DDR1 cell junctions, and inhibition of Src activity decreased Sh-DDR1-induced E-cadherin endocytosis. To characterise the molecular mechanisms, blocking integrin β1 decreased Src activity and E-cadherin junctional localisation in Sh-DDR1 cells. Photoconversion results showed that inhibition of Src activity rescued E-cadherin membrane stability and that inhibition of integrin β1-Src signalling decreased stress fibres and rescued E-cadherin membrane stability in Sh-DDR1 cells. Taken together, DDR1 stabilised membrane localisation of E-cadherin by inhibiting the integrin β1-Src-mediated clathrin-dependent endocytosis pathway.
Collapse
|
19
|
Rammal H, Saby C, Magnien K, Van-Gulick L, Garnotel R, Buache E, El Btaouri H, Jeannesson P, Morjani H. Discoidin Domain Receptors: Potential Actors and Targets in Cancer. Front Pharmacol 2016; 7:55. [PMID: 27014069 PMCID: PMC4789497 DOI: 10.3389/fphar.2016.00055] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/29/2016] [Indexed: 01/11/2023] Open
Abstract
The extracellular matrix critically controls cancer cell behavior by inducing several signaling pathways through cell membrane receptors. Besides conferring structural properties to tissues around the tumor, the extracellular matrix is able to regulate cell proliferation, survival, migration, and invasion. Among these receptors, the integrins family constitutes a major class of receptors that mediate cell interactions with extracellular matrix components. Twenty years ago, a new class of extracellular matrix receptors has been discovered. These tyrosine kinase receptors are the two discoidin domain receptors DDR1 and DDR2. DDR1 was first identified in the Dictyostelium discoideum and was shown to mediate cell aggregation. DDR2 shares highly conserved sequences with DDR1. Both receptors are activated upon binding to collagen, one of the most abundant proteins in extracellular matrix. While DDR2 can only be activated by fibrillar collagen, particularly types I and III, DDR1 is mostly activated by type I and IV collagens. In contrast with classical growth factor tyrosine kinase receptors which display a rapid and transient activation, DDR1 and DDR2 are unique in that they exhibit delayed and sustained receptor phosphorylation upon binding to collagen. Recent studies have reported differential expression and mutations of DDR1 and DDR2 in several cancer types and indicate clearly that these receptors have to be taken into account as new players in the different aspects of tumor progression, from non-malignant to highly malignant and invasive stages. This review will discuss the current knowledge on the role of DDR1 and DDR2 in malignant transformation, cell proliferation, epithelial to mesenchymal transition, migratory, and invasive processes, and finally the modulation of the response to chemotherapy. These new insights suggest that DDR1 and DDR2 are new potential targets in cancer therapy.
Collapse
Affiliation(s)
- Hassan Rammal
- Extracellular Matrix and Cellular Dynamics, Faculty of Pharmacy, MEDyC Centre National de la Recherche Scientifique UMR7369 Reims, France
| | - Charles Saby
- Extracellular Matrix and Cellular Dynamics, Faculty of Pharmacy, MEDyC Centre National de la Recherche Scientifique UMR7369 Reims, France
| | - Kevin Magnien
- Extracellular Matrix and Cellular Dynamics, Faculty of Pharmacy, MEDyC Centre National de la Recherche Scientifique UMR7369 Reims, France
| | - Laurence Van-Gulick
- Extracellular Matrix and Cellular Dynamics, Faculty of Pharmacy, MEDyC Centre National de la Recherche Scientifique UMR7369 Reims, France
| | - Roselyne Garnotel
- Extracellular Matrix and Cellular Dynamics, Faculty of Pharmacy, MEDyC Centre National de la Recherche Scientifique UMR7369 Reims, France
| | - Emilie Buache
- Extracellular Matrix and Cellular Dynamics, Faculty of Pharmacy, MEDyC Centre National de la Recherche Scientifique UMR7369 Reims, France
| | - Hassan El Btaouri
- Extracellular Matrix and Cellular Dynamics, Faculty of Pharmacy, MEDyC Centre National de la Recherche Scientifique UMR7369 Reims, France
| | - Pierre Jeannesson
- Extracellular Matrix and Cellular Dynamics, Faculty of Pharmacy, MEDyC Centre National de la Recherche Scientifique UMR7369 Reims, France
| | - Hamid Morjani
- Extracellular Matrix and Cellular Dynamics, Faculty of Pharmacy, MEDyC Centre National de la Recherche Scientifique UMR7369 Reims, France
| |
Collapse
|
20
|
Theocharis AD, Skandalis SS, Gialeli C, Karamanos NK. Extracellular matrix structure. Adv Drug Deliv Rev 2016; 97:4-27. [PMID: 26562801 DOI: 10.1016/j.addr.2015.11.001] [Citation(s) in RCA: 1382] [Impact Index Per Article: 172.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 12/12/2022]
Abstract
Extracellular matrix (ECM) is a non-cellular three-dimensional macromolecular network composed of collagens, proteoglycans/glycosaminoglycans, elastin, fibronectin, laminins, and several other glycoproteins. Matrix components bind each other as well as cell adhesion receptors forming a complex network into which cells reside in all tissues and organs. Cell surface receptors transduce signals into cells from ECM, which regulate diverse cellular functions, such as survival, growth, migration, and differentiation, and are vital for maintaining normal homeostasis. ECM is a highly dynamic structural network that continuously undergoes remodeling mediated by several matrix-degrading enzymes during normal and pathological conditions. Deregulation of ECM composition and structure is associated with the development and progression of several pathologic conditions. This article emphasizes in the complex ECM structure as to provide a better understanding of its dynamic structural and functional multipotency. Where relevant, the implication of the various families of ECM macromolecules in health and disease is also presented.
Collapse
Affiliation(s)
- Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece
| | - Chrysostomi Gialeli
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece; Division of Medical Protein Chemistry, Department of Translational Medicine Malmö, Lund University, S-20502 Malmö, Sweden
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece.
| |
Collapse
|
21
|
Hadzic E, Catillon M, Halavatyi A, Medves S, Van Troys M, Moes M, Baird MA, Davidson MW, Schaffner-Reckinger E, Ampe C, Friederich E. Delineating the Tes Interaction Site in Zyxin and Studying Cellular Effects of Its Disruption. PLoS One 2015; 10:e0140511. [PMID: 26509500 PMCID: PMC4624954 DOI: 10.1371/journal.pone.0140511] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 09/25/2015] [Indexed: 01/21/2023] Open
Abstract
Focal adhesions are integrin-based structures that link the actin cytoskeleton and the extracellular matrix. They play an important role in various cellular functions such as cell signaling, cell motility and cell shape. To ensure and fine tune these different cellular functions, adhesions are regulated by a large number of proteins. The LIM domain protein zyxin localizes to focal adhesions where it participates in the regulation of the actin cytoskeleton. Because of its interactions with a variety of binding partners, zyxin has been proposed to act as a molecular scaffold. Here, we studied the interaction of zyxin with such a partner: Tes. Similar to zyxin, Tes harbors three highly conserved LIM domains of which the LIM1 domain directly interacts with zyxin. Using different zyxin variants in pull-down assays and ectopic recruitment experiments, we identified the Tes binding site in zyxin and showed that four highly conserved amino acids are crucial for its interaction with Tes. Based upon these findings, we used a zyxin mutant defective in Tes-binding to assess the functional consequences of abrogating the zyxin-Tes interaction in focal adhesions. Performing fluorescence recovery after photobleaching, we showed that zyxin recruits Tes to focal adhesions and modulates its turnover in these structures. However, we also provide evidence for zyxin-independent localization of Tes to focal adhesions. Zyxin increases focal adhesion numbers and reduces focal adhesion lifetimes, but does so independent of Tes. Quantitative analysis showed that the loss of interaction between zyxin and Tes affects the process of cell spreading. We conclude that zyxin influences focal adhesion dynamics, that it recruits Tes and that this interaction is functional in regulating cell spreading.
Collapse
Affiliation(s)
- Ermin Hadzic
- Laboratory of Cytoskeleton and Cell Plasticity, Life Sciences Research Unit, University of Luxembourg, Luxemburg, Luxembourg
| | - Marie Catillon
- Laboratory of Cytoskeleton and Cell Plasticity, Life Sciences Research Unit, University of Luxembourg, Luxemburg, Luxembourg
| | - Aliaksandr Halavatyi
- Laboratory of Cytoskeleton and Cell Plasticity, Life Sciences Research Unit, University of Luxembourg, Luxemburg, Luxembourg
| | - Sandrine Medves
- Laboratory of Cytoskeleton and Cell Plasticity, Life Sciences Research Unit, University of Luxembourg, Luxemburg, Luxembourg
| | | | - Michèle Moes
- Laboratory of Cytoskeleton and Cell Plasticity, Life Sciences Research Unit, University of Luxembourg, Luxemburg, Luxembourg
| | - Michelle A. Baird
- National High Magnetic Field Laboratory and Department of Biological Science, The Florida State University, Tallahassee, Florida, United States of America
| | - Michael W. Davidson
- National High Magnetic Field Laboratory and Department of Biological Science, The Florida State University, Tallahassee, Florida, United States of America
| | - Elisabeth Schaffner-Reckinger
- Laboratory of Cytoskeleton and Cell Plasticity, Life Sciences Research Unit, University of Luxembourg, Luxemburg, Luxembourg
| | - Christophe Ampe
- Department of Biochemistry, Ghent University, Ghent, Belgium
- * E-mail:
| | - Evelyne Friederich
- Laboratory of Cytoskeleton and Cell Plasticity, Life Sciences Research Unit, University of Luxembourg, Luxemburg, Luxembourg
| |
Collapse
|
22
|
Juin A, Di Martino J, Leitinger B, Henriet E, Gary AS, Paysan L, Bomo J, Baffet G, Gauthier-Rouvière C, Rosenbaum J, Moreau V, Saltel F. Discoidin domain receptor 1 controls linear invadosome formation via a Cdc42-Tuba pathway. ACTA ACUST UNITED AC 2015; 207:517-33. [PMID: 25422375 PMCID: PMC4242841 DOI: 10.1083/jcb.201404079] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In tumor cells, the collagen receptor DDR1 collaborates with Cdc42 and its guanine exchange factor Tuba to promote linear invadosome formation and increase their matrix-invading activity. Accumulation of type I collagen fibrils in tumors is associated with an increased risk of metastasis. Invadosomes are F-actin structures able to degrade the extracellular matrix. We previously found that collagen I fibrils induced the formation of peculiar linear invadosomes in an unexpected integrin-independent manner. Here, we show that Discoidin Domain Receptor 1 (DDR1), a collagen receptor overexpressed in cancer, colocalizes with linear invadosomes in tumor cells and is required for their formation and matrix degradation ability. Unexpectedly, DDR1 kinase activity is not required for invadosome formation or activity, nor is Src tyrosine kinase. We show that the RhoGTPase Cdc42 is activated on collagen in a DDR1-dependent manner. Cdc42 and its specific guanine nucleotide-exchange factor (GEF), Tuba, localize to linear invadosomes, and both are required for linear invadosome formation. Finally, DDR1 depletion blocked cell invasion in a collagen gel. Altogether, our data uncover an important role for DDR1, acting through Tuba and Cdc42, in proteolysis-based cell invasion in a collagen-rich environment.
Collapse
Affiliation(s)
- Amélie Juin
- Institut National de la Santé et de la Recherche Médicale, U1053, F-33076 Bordeaux, France Université de Bordeaux, F-33076 Bordeaux, France
| | - Julie Di Martino
- Institut National de la Santé et de la Recherche Médicale, U1053, F-33076 Bordeaux, France Université de Bordeaux, F-33076 Bordeaux, France
| | - Birgit Leitinger
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, England, UK
| | - Elodie Henriet
- Institut National de la Santé et de la Recherche Médicale, U1053, F-33076 Bordeaux, France Université de Bordeaux, F-33076 Bordeaux, France
| | - Anne-Sophie Gary
- Institut National de la Santé et de la Recherche Médicale, U1053, F-33076 Bordeaux, France Université de Bordeaux, F-33076 Bordeaux, France
| | - Lisa Paysan
- Institut National de la Santé et de la Recherche Médicale, U1053, F-33076 Bordeaux, France Université de Bordeaux, F-33076 Bordeaux, France
| | - Jeremy Bomo
- Institut National de la Santé et de la Recherche Médicale, U1085, Institut de Recherche sur la Santé l'Environnement et le Travail (IRSET), Université de Rennes 1, 35043 Rennes, France
| | - Georges Baffet
- Institut National de la Santé et de la Recherche Médicale, U1085, Institut de Recherche sur la Santé l'Environnement et le Travail (IRSET), Université de Rennes 1, 35043 Rennes, France
| | - Cécile Gauthier-Rouvière
- Universités Montpellier 2 et 1, Centre de Recherche de Biochimie Macromoléculaire, Centre National de la Recherche Scientifique, UMR 5237, 34293 Montpellier, France
| | - Jean Rosenbaum
- Institut National de la Santé et de la Recherche Médicale, U1053, F-33076 Bordeaux, France Université de Bordeaux, F-33076 Bordeaux, France
| | - Violaine Moreau
- Institut National de la Santé et de la Recherche Médicale, U1053, F-33076 Bordeaux, France Université de Bordeaux, F-33076 Bordeaux, France
| | - Frédéric Saltel
- Institut National de la Santé et de la Recherche Médicale, U1053, F-33076 Bordeaux, France Université de Bordeaux, F-33076 Bordeaux, France
| |
Collapse
|
23
|
Reyes-Uribe E, Serna-Marquez N, Perez Salazar E. DDRs: receptors that mediate adhesion, migration and invasion in breast cancer cells. AIMS BIOPHYSICS 2015. [DOI: 10.3934/biophy.2015.3.303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
24
|
Leitinger B. Discoidin domain receptor functions in physiological and pathological conditions. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 310:39-87. [PMID: 24725424 DOI: 10.1016/b978-0-12-800180-6.00002-5] [Citation(s) in RCA: 253] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The discoidin domain receptors, DDR1 and DDR2, are nonintegrin collagen receptors that are members of the receptor tyrosine kinase family. Both DDRs bind a number of different collagen types and play important roles in embryo development. Dysregulated DDR function is associated with progression of various human diseases, including fibrosis, arthritis, and cancer. By interacting with key components of the extracellular matrix and displaying distinct activation kinetics, the DDRs form a unique subfamily of receptor tyrosine kinases. DDR-facilitated cellular functions include cell migration, cell survival, proliferation, and differentiation, as well as remodeling of extracellular matrices. This review summarizes the current knowledge of DDR-ligand interactions, DDR-initiated signal pathways and the molecular mechanisms that regulate receptor function. Also discussed are the roles of DDRs in development and disease progression.
Collapse
Affiliation(s)
- Birgit Leitinger
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| |
Collapse
|
25
|
Ghosh S, Ashcraft K, Jahid MJ, April C, Ghajar CM, Ruan J, Wang H, Foster M, Hughes DC, Ramirez AG, Huang T, Fan JB, Hu Y, Li R. Regulation of adipose oestrogen output by mechanical stress. Nat Commun 2013; 4:1821. [PMID: 23652009 DOI: 10.1038/ncomms2794] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 03/22/2013] [Indexed: 01/24/2023] Open
Abstract
Adipose stromal cells are the primary source of local oestrogens in adipose tissue, aberrant production of which promotes oestrogen receptor-positive breast cancer. Here we show that extracellular matrix compliance and cell contractility are two opposing determinants for oestrogen output of adipose stromal cells. Using synthetic extracellular matrix and elastomeric micropost arrays with tunable rigidity, we find that increasing matrix compliance induces transcription of aromatase, a rate-limiting enzyme in oestrogen biosynthesis. This mechanical cue is transduced sequentially by discoidin domain receptor 1, c-Jun N-terminal kinase 1, and phosphorylated JunB, which binds to and activates two breast cancer-associated aromatase promoters. In contrast, elevated cell contractility due to actin stress fibre formation dampens aromatase transcription. Mechanically stimulated stromal oestrogen production enhances oestrogen-dependent transcription in oestrogen receptor-positive tumour cells and promotes their growth. This novel mechanotransduction pathway underlies communications between extracellular matrix, stromal hormone output, and cancer cell growth within the same microenvironment.
Collapse
Affiliation(s)
- Sagar Ghosh
- Department of Molecular Medicine/Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Discoidin domain receptors in disease. Matrix Biol 2013; 34:185-92. [PMID: 24361528 DOI: 10.1016/j.matbio.2013.12.002] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 12/05/2013] [Accepted: 12/05/2013] [Indexed: 12/17/2022]
Abstract
Discoidin domain receptors, DDR1 and DDR2, lie at the intersection of two large receptor families, namely the extracellular matrix and tyrosine kinase receptors. As such, DDRs are uniquely positioned to function as sensors for extracellular matrix and to regulate a wide range of cell functions from migration and proliferation to cytokine secretion and extracellular matrix homeostasis/remodeling. While activation of DDRs by extracellular matrix collagens is required for normal development and tissue homeostasis, aberrant activation of these receptors following injury or in disease is detrimental. The availability of mice lacking DDRs has enabled us to identify key roles played by these receptors in disease initiation and progression. DDR1 promotes inflammation in atherosclerosis, lung fibrosis and kidney injury, while DDR2 contributes to osteoarthritis. Furthermore, both DDRs have been implicated in cancer progression. Yet the mechanisms whereby DDRs contribute to disease progression are poorly understood. In this review we highlight the mechanisms whereby DDRs regulate two important processes, namely inflammation and tissue fibrosis. In addition, we discuss the challenges of targeting DDRs in disease. Selective targeting of these receptors requires understanding of how they interact with and are activated by extracellular matrix, and whether their cellular function is dependent on or independent of receptor kinase activity.
Collapse
|
27
|
Staudinger LA, Spano SJ, Lee W, Coelho N, Rajshankar D, Bendeck MP, Moriarty T, McCulloch CA. Interactions between the discoidin domain receptor 1 and β1 integrin regulate attachment to collagen. Biol Open 2013; 2:1148-59. [PMID: 24244851 PMCID: PMC3828761 DOI: 10.1242/bio.20135090] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 08/21/2013] [Indexed: 12/15/2022] Open
Abstract
Collagen degradation by phagocytosis is essential for physiological collagen turnover and connective tissue homeostasis. The rate limiting step of phagocytosis is the binding of specific adhesion receptors, which include the integrins and discoidin domain receptors (DDR), to fibrillar collagen. While previous data suggest that these two receptors interact, the functional nature of these interactions is not defined. In mouse and human fibroblasts we examined the effects of DDR1 knockdown and over-expression on β1 integrin subunit function. DDR1 expression levels were positively associated with enhanced contraction of floating and attached collagen gels, increased collagen binding and increased collagen remodeling. In DDR1 over-expressing cells compared with control cells, there were increased numbers, area and length of focal adhesions immunostained for talin, paxillin, vinculin and activated β1 integrin. After treatment with the integrin-cleaving protease jararhagin, in comparison to controls, DDR1 over-expressing cells exhibited increased β1 integrin cleavage at the cell membrane, indicating that DDR1 over-expression affected the access and susceptibility of cell-surface β1 integrin to the protease. DDR1 over-expression was associated with increased glycosylation of the β1 integrin subunit, which when blocked by deoxymannojirimycin, reduced collagen binding. Collectively these data indicate that DDR1 regulates β1 integrin interactions with fibrillar collagen, which positively impacts the binding step of collagen phagocytosis and collagen remodeling.
Collapse
Affiliation(s)
- Lisa A Staudinger
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto , Toronto, ON M5S 3E2 , Canada
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Collagen XV inhibits epithelial to mesenchymal transition in pancreatic adenocarcinoma cells. PLoS One 2013; 8:e72250. [PMID: 23991074 PMCID: PMC3750028 DOI: 10.1371/journal.pone.0072250] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 07/12/2013] [Indexed: 01/13/2023] Open
Abstract
Collagen XV (COLXV) is a secreted non-fibrillar collagen found within basement membrane (BM) zones of the extracellular matrix (ECM). Its ability to alter cellular growth in vitro and to reduce tumor burden and increase survival in vivo support a role as a tumor suppressor. Loss of COLXV during the progression of several aggressive cancers precedes basement membrane invasion and metastasis. The resultant lack of COLXV subjacent to the basement membrane and subsequent loss of its interactions with other proteins in this zone may directly impact tumor progression. Here we show that COLXV significantly reduces invasion of pancreatic adenocarcinoma cells through a collagen I (COLI) matrix. Moreover, we demonstrate that epithelial to mesenchymal transition (EMT) in these cells, which is recapitulated in vitro by cell scattering on a COLI substrate, is inhibited by over-expression of COLXV. We identify critical collagen-binding surface receptors on the tumor cells, including the discoidin domain receptor 1 (DDR1) and E-Cadherin (E-Cad), which interact with COLXV and appear to mediate its function. In the presence of COLXV, the intracellular redistribution of E-Cad from the cell periphery, which is associated with COLI-activated EMT, is inhibited and concurrently, DDR1 signaling is suppressed. Furthermore, continuous exposure of the pancreatic adenocarcinoma cells to high levels of COLXV suppresses endogenous levels of N-Cadherin (N-Cad). These data reveal a novel mechanism whereby COLXV can function as a tumor suppressor in the basement membrane zone.
Collapse
|
29
|
Fu HL, Valiathan RR, Arkwright R, Sohail A, Mihai C, Kumarasiri M, Mahasenan KV, Mobashery S, Huang P, Agarwal G, Fridman R. Discoidin domain receptors: unique receptor tyrosine kinases in collagen-mediated signaling. J Biol Chem 2013; 288:7430-7437. [PMID: 23335507 DOI: 10.1074/jbc.r112.444158] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The discoidin domain receptors (DDRs) are receptor tyrosine kinases that recognize collagens as their ligands. DDRs display unique structural features and distinctive activation kinetics, which set them apart from other members of the kinase superfamily. DDRs regulate cell-collagen interactions in normal and pathological conditions and thus are emerging as major sensors of collagen matrices and potential novel therapeutic targets. New structural and biological information has shed light on the molecular mechanisms that regulate DDR signaling, turnover, and function. This minireview provides an overview of these areas of DDR research with the goal of fostering further investigation of these intriguing and unique receptors.
Collapse
Affiliation(s)
- Hsueh-Liang Fu
- Department of Pathology, School of Medicine, Wayne State University, Detroit, Michigan 48201
| | - Rajeshwari R Valiathan
- Department of Pathology, School of Medicine, Wayne State University, Detroit, Michigan 48201
| | - Richard Arkwright
- Department of Pathology, School of Medicine, Wayne State University, Detroit, Michigan 48201
| | - Anjum Sohail
- Department of Pathology, School of Medicine, Wayne State University, Detroit, Michigan 48201
| | - Cosmin Mihai
- Davis Heart and Lung Research Institute and Biomedical Engineering Department, Ohio State University, Columbus, Ohio 43210
| | - Malika Kumarasiri
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556
| | - Kiran V Mahasenan
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556
| | - Paul Huang
- Institute of Cancer Research, Chester Beatty Laboratories, London SW3 6JB, United Kingdom
| | - Gunjan Agarwal
- Davis Heart and Lung Research Institute and Biomedical Engineering Department, Ohio State University, Columbus, Ohio 43210
| | - Rafael Fridman
- Department of Pathology, School of Medicine, Wayne State University, Detroit, Michigan 48201.
| |
Collapse
|
30
|
Xu H, Bihan D, Chang F, Huang PH, Farndale RW, Leitinger B. Discoidin domain receptors promote α1β1- and α2β1-integrin mediated cell adhesion to collagen by enhancing integrin activation. PLoS One 2012; 7:e52209. [PMID: 23284937 PMCID: PMC3527415 DOI: 10.1371/journal.pone.0052209] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 11/09/2012] [Indexed: 11/18/2022] Open
Abstract
The discoidin domain receptors, DDR1 and DDR2, are receptor tyrosine kinases that bind to and are activated by collagens. Similar to collagen-binding β1 integrins, the DDRs bind to specific motifs within the collagen triple helix. However, these two types of collagen receptors recognize distinct collagen sequences. While GVMGFO (O is hydroxyproline) functions as a major DDR binding motif in fibrillar collagens, integrins bind to sequences containing Gxx'GEx". The DDRs are thought to regulate cell adhesion, but their roles have hitherto only been studied indirectly. In this study we used synthetic triple-helical collagen-derived peptides that incorporate either the DDR-selective GVMGFO motif or integrin-selective motifs, such as GxOGER and GLOGEN, in order to selectively target either type of receptor and resolve their contributions to cell adhesion. Our data using HEK293 cells show that while cell adhesion to collagen I was completely inhibited by anti-integrin blocking antibodies, the DDRs could mediate cell attachment to the GVMGFO motif in an integrin-independent manner. Cell binding to GVMGFO was independent of DDR receptor signalling and occurred with limited cell spreading, indicating that the DDRs do not mediate firm adhesion. However, blocking the interaction of DDR-expressing cells with collagen I via the GVMGFO site diminished cell adhesion, suggesting that the DDRs positively modulate integrin-mediated cell adhesion. Indeed, overexpression of the DDRs or activation of the DDRs by the GVMGFO ligand promoted α1β1 and α2β1 integrin-mediated cell adhesion to medium- and low-affinity integrin ligands without regulating the cell surface expression levels of α1β1 or α2β1. Our data thus demonstrate an adhesion-promoting role of the DDRs, whereby overexpression and/or activation of the DDRs leads to enhanced integrin-mediated cell adhesion as a result of higher integrin activation state.
Collapse
Affiliation(s)
- Huifang Xu
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Dominique Bihan
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Francis Chang
- Division of Cancer Biology, Institute of Cancer Research, London, United Kingdom
| | - Paul H. Huang
- Division of Cancer Biology, Institute of Cancer Research, London, United Kingdom
| | - Richard W. Farndale
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Birgit Leitinger
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
31
|
Yeh YC, Lin HH, Tang MJ. A tale of two collagen receptors, integrin β1 and discoidin domain receptor 1, in epithelial cell differentiation. Am J Physiol Cell Physiol 2012; 303:C1207-17. [DOI: 10.1152/ajpcell.00253.2012] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
As increase in collagen deposition is no longer taken as simply a consequence but, rather, an inducer of disease progression; therefore, the understanding of collagen signal transduction is fundamentally important. Cells contain at least two types of collagen receptors: integrins and discoidin domain receptors (DDRs). The integrin heterodimers α1β1, α2β1, α10β1, and α11β1 are recognized as the non-tyrosine kinase collagen receptors. DDR1 and 2, the tyrosine kinase receptors of collagen, are specifically expressed in epithelium and mesenchyme, respectively. While integrin β1 and DDR1 are both required for cell adhesion on collagen, their roles in epithelial cell differentiation during development and disease progression seem to counteract each other, with integrin β1 favoring epithelium mesenchyme transition (EMT) and DDR1 inducing epithelial cell differentiation. The in vitro evidence shows that the integrin β1 and DDR1 exert opposing actions in regulation of membrane stability of E-cadherin, which itself is a critical regulator of epithelial cell differentiation. Here, we review the functional roles of integrin β1 and DDR1 in regulation of epithelial cell differentiation during development and disease progression, and explore the underlining mechanisms regarding to the regulation of membrane stability of E-cadherin.
Collapse
Affiliation(s)
- Yi-Chun Yeh
- Department of Physiology, National Cheng Kung University Medicine College, Tainan, Taiwan; and
| | - Hsi-Hui Lin
- Department of Physiology, National Cheng Kung University Medicine College, Tainan, Taiwan; and
| | - Ming-Jer Tang
- Department of Physiology, National Cheng Kung University Medicine College, Tainan, Taiwan; and
- Center for Gene Regulation and Signal Transduction, National Cheng Kung University Medicine College, Tainan, Taiwan
| |
Collapse
|
32
|
Herrera-Herrera ML, Quezada-Calvillo R. DDR2 plays a role in fibroblast migration independent of adhesion ligand and collagen activated DDR2 tyrosine kinase. Biochem Biophys Res Commun 2012; 429:39-44. [PMID: 23131558 DOI: 10.1016/j.bbrc.2012.10.103] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 10/17/2012] [Indexed: 12/22/2022]
Abstract
Discoidin domain receptor-2 (DDR2) is a cell surface tyrosine kinase receptor that can be activated by soluble collagen and has been implicated in diverse physiological functions including organism growth and wound repair. In the current studies, we used fibronectin and collagen-coated 2D surfaces and collagen matrices in combination with siRNA technology to investigate the role of DDR2 in a range of fibroblast motile activities. Silencing DDR2 with siRNA inhibited cell spreading and migration, and similar inhibition occurred regardless whether cells were interacting with fibronectin or collagen surfaces. Under the assay conditions used, DDR2 tyrosine kinase activation was not observed unless soluble collagen was added to the incubation medium. Finally silencing DDR2 also inhibited human fibroblast migration in 3D collagen matrices but had no effect on 3D collagen matrix remodeling and contraction. Taken together, our findings suggest that DDR2 is required for normal fibroblast spreading and migration independent of adhesion ligand and collagen activation of DDR2 tyrosine kinase.
Collapse
|
33
|
Valiathan RR, Marco M, Leitinger B, Kleer CG, Fridman R. Discoidin domain receptor tyrosine kinases: new players in cancer progression. Cancer Metastasis Rev 2012; 31:295-321. [PMID: 22366781 DOI: 10.1007/s10555-012-9346-z] [Citation(s) in RCA: 282] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Almost all human cancers display dysregulated expression and/or function of one or more receptor tyrosine kinases (RTKs). The strong causative association between altered RTK function and cancer progression has been translated into novel therapeutic strategies that target these cell surface receptors in cancer. Yet, the full spectrum of RTKs that may alter the oncogenic process is not completely understood. Accumulating evidence suggests that a unique set of RTKs known as the discoidin domain receptors (DDRs) play a key role in cancer progression by regulating the interactions of tumor cells with their surrounding collagen matrix. The DDRs are the only RTKs that specifically bind to and are activated by collagen. DDRs control cell and tissue homeostasis by acting as collagen sensors, transducing signals that regulate cell polarity, tissue morphogenesis, and cell differentiation. In cancer, DDRs are hijacked by tumor cells to disrupt normal cell-matrix communication and initiate pro-migratory and pro-invasive programs. Importantly, several cancer types exhibit DDR mutations, which are thought to alter receptor function and contribute to cancer progression. Other evidence suggests that the actions of DDRs in cancer are complex, either promoting or suppressing tumor cell behavior in a DDR type/isoform specific- and context-dependent manner. Thus, there is still a considerable gap in our knowledge of DDR actions in cancer tissues. This review summarizes and discusses the current knowledge on DDR expression and function in cancer. It is hoped that this effort will encourage more research into these poorly understood but unique RTKs, which have the potential of becoming novel therapeutic targets in cancer.
Collapse
Affiliation(s)
- Rajeshwari R Valiathan
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | |
Collapse
|
34
|
Side population cells in human gallbladder cancer cell line GBC-SD regulated by TGF-β-induced epithelial-mesenchymal transition. ACTA ACUST UNITED AC 2011; 31:749-755. [DOI: 10.1007/s11596-011-0671-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Indexed: 12/31/2022]
|
35
|
Abstract
Collagen, the most abundant protein in animals, is a key component of extracellular matrices. Not only do collagens provide essential structural support for connective tissues, but they are also intimately involved in controlling a spectrum of cellular functions such as growth, differentiation, and morphogenesis. All collagens possess triple-helical regions through which they interact with a host of other proteins including cell surface receptors. A structurally diverse group of transmembrane receptors mediates the recognition of the collagen triple helix: integrins, discoidin domain receptors, glycoprotein VI, and leukocyte-associated immunoglobulin-like receptor-1. These collagen receptors regulate a wide range of behaviors including cell adhesion and migration, hemostasis, and immune function. Here these collagen receptors are discussed in terms of their molecular basis of collagen recognition, their signaling and developmental functions, and their roles in disease.
Collapse
Affiliation(s)
- Birgit Leitinger
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, United Kingdom.
| |
Collapse
|
36
|
Xu B, Bhullar RP. Regulation of Rac1 and Cdc42 activation in thrombin- and collagen-stimulated CHRF-288-11 cells. Mol Cell Biochem 2011; 353:73-9. [DOI: 10.1007/s11010-011-0776-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Accepted: 03/07/2011] [Indexed: 11/30/2022]
|
37
|
Yeh YC, Wu CC, Wang YK, Tang MJ. DDR1 triggers epithelial cell differentiation by promoting cell adhesion through stabilization of E-cadherin. Mol Biol Cell 2011; 22:940-53. [PMID: 21289093 PMCID: PMC3069019 DOI: 10.1091/mbc.e10-08-0678] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Discoidin domain receptor 1 (DDR1) promotes cell differentiation through the increase of E-cadherin-mediated cell-cell contact. Life cell imaging with E-cadherin conjugated with Eos fluorescence protein showed that DDR1 stabilizes membrane-bound E-cadherin and the inactivation of Cdc42 mediates DDR1-regulated cell adhesion and differentiation. Discoidin domain receptor 1 (DDR1) promotes E-cadherin–mediated adhesion. The underlying mechanism and its significance, however, have not been elucidated. Here we show that DDR1 overexpression augmented, whereas dominant negative mutant (DN-DDR1) or knockdown of DDR1 inhibited E-cadherin localized in cell-cell junctions in epithelial cells. DDR1 changed the localization and abundance of E-cadherin, as well as epithelial plasticity, as manifested by enhancement of microvilli formation and alteration of cytoskeletal organization. DDR1 also reduced protein abundance of mesenchymal markers, whereas DN-DDR1 and sh-DDR1 showed opposite effects. These results suggest that expression of DDR1 increases epithelial plasticity. Expression of DDR1 augmented E-cadherin protein levels by decreasing its degradation rate. Photobleaching and photoconversion of E-cadherin conjugated with Eos fluorescence protein demonstrated that DDR1 increased the stability of E-cadherin on the cell membrane, whereas sh-DDR1 decreased it. Pull-down assay and expression of constitutively active or dominant-negative Cdc42 showed that DDR1 stabilized E-cadherin through inactivation of Cdc42. Altogether, our results show that DDR1 promotes cell-cell adhesion and differentiation through stabilization of E-cadherin, which is mediated by Cdc42 inactivation.
Collapse
Affiliation(s)
- Yi-Chun Yeh
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan, Taiwan
| | | | | | | |
Collapse
|
38
|
Yeh YC, Wei WC, Wang YK, Lin SC, Sung JM, Tang MJ. Transforming growth factor-{beta}1 induces Smad3-dependent {beta}1 integrin gene expression in epithelial-to-mesenchymal transition during chronic tubulointerstitial fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:1743-54. [PMID: 20709799 DOI: 10.2353/ajpath.2010.091183] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Transforming growth factor-β1 (TGF-β1)-induced epithelial-to-mesenchymal transition (EMT) contributes to the pathophysiological development of kidney fibrosis. Although it was reported that TGF-β1 enhances β(1) integrin levels in NMuMG cells, the detailed molecular mechanisms underlying TGF-β1-induced β(1) integrin gene expression and the role of β(1) integrin during EMT in the renal system are still unclear. In this study, we examined the role of β(1) integrin in TGF-β1-induced EMT both in vitro and in vivo. TGF-β1-induced augmentation of β(1) integrin expression was required for EMT in several epithelial cell lines, and knockdown of Smad3 inhibited TGF-β1-induced augmentation of β(1) integrin. TGF-β1 triggered β(1) integrin gene promoter activity as assessed by luciferase activity assay. Both knockdown of Smad3 and mutation of the Smad-binding element to block binding to the β(1) integrin promoter markedly reduced TGF-β1-induced β(1) integrin promoter activity. Chromatin immunoprecipitation assay showed that TGF-β1 enhanced Smad3 binding to the β(1) integrin promoter. Furthermore, induction of unilateral ureteral obstruction triggered increases of β(1) integrin in both renal epithelial and interstitial cells. In human kidney with chronic tubulointerstitial fibrosis, we also found a concomitant increase of β(1) integrin and α-smooth muscle actin in tubule epithelia. Blockade of β(1) integrin signaling dampened the progression of fibrosis. Taken together, β(1) integrin mediates EMT and subsequent tubulointerstitutial fibrosis, suggesting that inhibition of β(1) integrin is a possible therapeutic target for prevention of renal fibrosis.
Collapse
Affiliation(s)
- Yi-Chun Yeh
- tDepartment of Medicine, Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Skeleton-Joint Research Center, Tainan 70101, Taiwan
| | | | | | | | | | | |
Collapse
|
39
|
Franco C, Ahmad PJ, Hou G, Wong E, Bendeck MP. Increased cell and matrix accumulation during atherogenesis in mice with vessel wall-specific deletion of discoidin domain receptor 1. Circ Res 2010; 106:1775-83. [PMID: 20448217 DOI: 10.1161/circresaha.109.213637] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Discoidin domain receptor (DDR)1 is a collagen receptor expressed on both smooth muscle cells (SMCs) and macrophages, where it plays important roles regulating cell and matrix accumulation during atherogenesis. Systemic deletion of DDR1 resulted in attenuated plaque growth but accelerated matrix accumulation in LDLR-deficient mice. Deletion of DDR1 solely on bone marrow-derived cells resulted in decreased macrophage accumulation and plaque growth but no change in matrix accumulation. OBJECTIVE These findings led us to hypothesize that accelerated matrix accumulation was attributable to the increased synthetic ability of Ddr1(-/-) resident vascular wall SMCs. METHODS AND RESULTS We used bone marrow transplantation to generate chimeric mice and investigate the role of SMC DDR1 during atherogenesis. Mice with deficiency of DDR1 in vessel wall-derived cells (Ddr1(+/+-->-/-)) or control mice (Ddr1(+/+-->+/+)) were fed an atherogenic diet for 12 weeks. We observed a 3.8-fold increase in the size of aortic sinus plaques in Ddr1(+/+-->-/-) compared to Ddr1(+/+-->+/+) mice. This was attributed to pronounced accumulation of collagen, elastin, proteoglycans, and fibronectin and resulted in a thickened fibrous cap. The enhanced matrix accumulation decreased the proportion of plaque area occupied by cells but was associated with a shift in the cellular composition of the lesions toward increased numbers of vessel wall-derived SMCs compared to bone marrow-derived macrophages. In vitro studies confirmed that Ddr1(-/-) SMCs expressed more matrix, proliferated more, and migrated farther than Ddr1(+/+) SMCs. CONCLUSIONS DDR1 expression on resident vessel wall SMCs limits proliferation, migration and matrix accumulation during atherogenesis.
Collapse
Affiliation(s)
- Christopher Franco
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, Room 6213, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | | | | | | | | |
Collapse
|
40
|
Eswaramoorthy R, Wang CK, Chen WC, Tang MJ, Ho ML, Hwang CC, Wang HM, Wang CZ. DDR1 regulates the stabilization of cell surface E-cadherin and E-cadherin-mediated cell aggregation. J Cell Physiol 2010; 224:387-97. [PMID: 20432435 DOI: 10.1002/jcp.22134] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
41
|
Roig B, Franco-Pons N, Martorell L, Tomàs J, Vogel WF, Vilella E. Expression of the tyrosine kinase discoidin domain receptor 1 (DDR1) in human central nervous system myelin. Brain Res 2010; 1336:22-9. [PMID: 20380825 DOI: 10.1016/j.brainres.2010.03.099] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 03/26/2010] [Accepted: 03/29/2010] [Indexed: 12/11/2022]
Abstract
During development of the mouse brain, the protein kinase discoidin domain receptor 1 (DDR1) is present prenatally in neurons of the proliferative areas, and postnatally, DDR1 expression is no longer detected in neurons, but a spatial-temporal expression pattern in oligodendrocytes that overlaps with the dynamics of the myelination process is detected. Notably, oligodendrocytic DDR1 expression is upregulated in mice during experimentally induced remyelination. Recently, we demonstrated that DDR1 expression is high in human brain and that there is an association between the gene and schizophrenia in a case-control study. However, data regarding expression of DDR1 in the human brain are scarce. Here, we describe the expression pattern of DDR1 in the human adult cerebral cortex. Using several immunohistological techniques and in situ hybridization, we identified DDR1 in the following: a) myelin, b) capillary endothelial cells in the gray as well as white matter, and c) in the soma of some oligodendrocytes and astrocytes in the white matter. The most important overall finding in this study was that DDR1 is present in myelin and is expressed by oligodendrocyte cells. We detected the presence of DDR1 mRNA and protein in myelin and observed that DDR1 co-localized with the classical myelin basic protein (MBP). Moreover, we found a strong positive correlation between expression levels of DDR1 and two myelin-associated genes, myelin-associated glycoprotein (MAG) and oligodendrocyte transcription factor 2 (OLIG2). These observations suggest that DDR1 could be an important constituent of myelin. Because defects in myelination are linked to several mental disorders such as schizophrenia, the function of DDR1 in the process of myelination warrants further investigation.
Collapse
Affiliation(s)
- Bàrbara Roig
- Hospital Psiquiàtric Universitari Institut Pere Mata, IISPV, Universitat Rovira i Virgili, Reus, Spain
| | | | | | | | | | | |
Collapse
|
42
|
Maillet M, Lynch JM, Sanna B, York AJ, Zheng Y, Molkentin JD. Cdc42 is an antihypertrophic molecular switch in the mouse heart. J Clin Invest 2009; 119:3079-88. [PMID: 19741299 DOI: 10.1172/jci37694] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2008] [Accepted: 07/08/2009] [Indexed: 01/19/2023] Open
Abstract
To improve contractile function, the myocardium undergoes hypertrophic growth without myocyte proliferation in response to both pathologic and physiologic stimulation. Various membrane-bound receptors and intermediate signal transduction pathways regulate the induction of cardiac hypertrophy, but the cardioprotective regulatory pathways or effectors that antagonize cardiac hypertrophy remain poorly understood. Here we identify the small GTPase Cdc42 as a signaling intermediate that restrained the cardiac growth response to physiologic and pathologic stimuli. Cdc42 was specifically activated in the heart after pressure overload and in cultured cardiomyocytes by multiple agonists. Mice with a heart-specific deletion of Cdc42 developed greater cardiac hypertrophy at 2 and 8 weeks of stimulation and transitioned more quickly into heart failure than did wild-type controls. These mice also displayed greater cardiac hypertrophy in response to neuroendocrine agonist infusion for 2 weeks and, more remarkably, enhanced exercise-induced hypertrophy and sudden death. These pathologies were associated with an inability to activate JNK following stimulation through a MEKK1/MKK4/MKK7 pathway, resulting in greater cardiac nuclear factor of activated T cells (NFAT) activity. Restoration of cardiac JNK signaling with an Mkk7 heart-specific transgene reversed the enhanced growth effect. These results identify what we believe to be a novel antihypertrophic and protective cardiac signaling pathway, whereby Cdc42-dependent JNK activation antagonizes calcineurin-NFAT activity to reduce hypertrophy and prevent transition to heart failure.
Collapse
Affiliation(s)
- Marjorie Maillet
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, University of Cincinnati, and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | | | | | | | | |
Collapse
|
43
|
Wang CZ, Yeh YC, Tang MJ. DDR1/E-cadherin complex regulates the activation of DDR1 and cell spreading. Am J Physiol Cell Physiol 2009; 297:C419-29. [PMID: 19474292 DOI: 10.1152/ajpcell.00101.2009] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Discoidin domain receptors (DDRs) 1 and 2, collagen receptors, regulate cell adhesion and a broad range of cell behavior. Their adhesion-dependent regulation of signaling associated with adhesion proteins has not been elucidated. We report a novel mechanism: the cross talk of DDR1 and E-cadherin negatively and adhesion dependently regulated both DDR1 activity and DDR1-suppressed cell spreading. E-cadherin forms complexes with both DDR1 isoforms (a and b). E-cadherin regulates DDR1 activity associated with the cell-junction complexes formed between DDR1 and E-cadherin. These complexes are formed independently of DDR1 activation and of beta-catenin and p120-catenin binding to E-cadherin; they are ubiquitous in epithelial cells. Small interfering RNA-mediated gene silencing of E-cadherin restores both DDR1 activity and DDR1-suppressed cell spreading and increases the apically and basally located DDR1 in E-cadherin-null cells. We conclude that E-cadherin-mediated adhesions decrease DDR1 activity, which subsequently eliminates DDR1-suppressed cell spreading, by sequestering DDR1 to cell junctions, which prevents its contact with collagen ligand.
Collapse
Affiliation(s)
- Chau-Zen Wang
- Department of Physiology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | |
Collapse
|
44
|
Huang Y, Arora P, McCulloch CA, Vogel WF. The collagen receptor DDR1 regulates cell spreading and motility by associating with myosin IIA. J Cell Sci 2009; 122:1637-46. [PMID: 19401332 DOI: 10.1242/jcs.046219] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The spreading and migration of cells on adhesive substrates is regulated by the counterbalance of contractile and protrusive forces. Non-muscle myosin IIA, an ubiquitously expressed contractile protein and enzyme, is implicated in the regulation of cell spreading and directional migration in response to various stimuli. Here we show that discoidin domain receptor 1 (DDR1), a tyrosine kinase receptor activated by type I collagen, associates with the non-muscle myosin IIA heavy chain (NMHC-IIA) upon ligand stimulation. An association was also indicated by coimmunoprecipitation of NMHC-IIA with full-length DDR1, but not with the truncated DDR1d-isoform lacking the kinase domain. DDR1 was important for assembly of NMHC-IIA into filaments on cells plated on collagen. DDR1 expression inhibited cell spreading over collagen but promoted cell migration. By contrast, blockade of non-muscle myosin II activity by blebbistatin enhanced cell spreading but inhibited migration over collagen. We propose that myosin and DDR1 impact cell spreading and migration by regulating adhesive contacts with collagen.
Collapse
Affiliation(s)
- Yun Huang
- Department of Laboratory Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | | | | | |
Collapse
|