1
|
Zhang S, Guo Y, Lu Y, Liu F, Heng BC, Deng X. The considerations on selecting the appropriate decellularized ECM for specific regeneration demands. Mater Today Bio 2024; 29:101301. [PMID: 39498148 PMCID: PMC11532911 DOI: 10.1016/j.mtbio.2024.101301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024] Open
Abstract
An ideal biomaterial should create a customized tissue-specific microenvironment that can facilitate and guide the tissue repair process. Due to its good biocompatibility and similar biochemical properties to native tissues, decellularized extracellular matrix (dECM) generally yields enhanced regenerative outcomes, with improved morphological and functional recovery. By utilizing various decellularization techniques and post-processing protocols, dECM can be flexibly prepared in different states from various sources, with specifically customized physicochemical properties for different tissues. To initiate a well-orchestrated tissue-regenerative response, dECM exerts multiple effects at the wound site by activating various overlapping signaling pathways to promote cell adhesion, proliferation, and differentiation, as well as suppressing inflammation via modulation of various immune cells, including macrophages, T cells, and mastocytes. Functional tissue repair is likely the main aim when employing the optimized dECM biomaterials. Here, we review the current applications of different kinds of dECMs in an attempt to improve the efficiency of tissue regeneration, highlighting key considerations on developing dECM for specific tissue engineering applications.
Collapse
Affiliation(s)
- Shihan Zhang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Yaru Guo
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Yixuan Lu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Fangyong Liu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Boon Chin Heng
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- NMPA Key Laboratory for Dental Materials, Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| |
Collapse
|
2
|
Angeli E, Jordan M, Otto M, Stojanović SD, Karsdal M, Bauersachs J, Thum T, Fiedler J, Genovese F. The role of fibrosis in cardiomyopathies: An opportunity to develop novel biomarkers of disease activity. Matrix Biol 2024; 128:65-78. [PMID: 38423395 DOI: 10.1016/j.matbio.2024.02.008] [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: 10/16/2023] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 03/02/2024]
Abstract
Cardiomyopathies encompass a spectrum of heart disorders with diverse causes and presentations. Fibrosis stands out as a shared hallmark among various cardiomyopathies, reflecting a common thread in their pathogenesis. This prevalent fibrotic response is intricately linked to the consequences of dysregulated extracellular matrix (ECM) remodeling, emphasizing its significance in the development and progression the disease. This review explores the ECM involvement in various cardiomyopathies and its impact on myocardial stiffness and fibrosis. Additionally, we discuss the potential of ECM fragments as early diagnosis, prognosis, and risk stratification. Biomarkers deriving from turnover of collagens and other ECM proteins hold promise in clinical applications. We outline current clinical management, future directions, and the potential for personalized ECM-targeted therapies with specific focus on microRNAs. In summary, this review examines the role of the fibrosis in cardiomyopathies, highlighting the potential of ECM-derived biomarkers in improving disease management with implications for precision medicine.
Collapse
Affiliation(s)
- Elisavet Angeli
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; Nordic Bioscience A/S, Herlev, Denmark.
| | - Maria Jordan
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Federal Republic of Germany; Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Hanover, Federal Republic of Germany
| | - Mandy Otto
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Federal Republic of Germany; Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Hanover, Federal Republic of Germany
| | - Stevan D Stojanović
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Federal Republic of Germany; Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Federal Republic of Germany
| | | | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Federal Republic of Germany
| | - Thomas Thum
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Federal Republic of Germany; Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Hanover, Federal Republic of Germany; Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Federal Republic of Germany
| | - Jan Fiedler
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Federal Republic of Germany; Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Hanover, Federal Republic of Germany
| | - Federica Genovese
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
Xu H, Tan M, Hou GQ, Sang YZ, Lin L, Gan XC, Cao X, Liu AD. Blockade of DDR1/PYK2/ERK signaling suggesting SH2 superbinder as a novel autophagy inhibitor for pancreatic cancer. Cell Death Dis 2023; 14:811. [PMID: 38071340 PMCID: PMC10710504 DOI: 10.1038/s41419-023-06344-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 11/12/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023]
Abstract
Pancreatic cancer is highly lethal, of which 90% is pancreatic ductal adenocarcinoma (PDAC), with a 5-year survival rate of less than 12%, lacking effective treatment options and late diagnosis. Furthermore, the tumors show an intense resistance to cytotoxic chemotherapies. As autophagy is elevated in PDAC, targeting the autophagic pathway is regarded as a promising strategy for cancer treatment. Immunofluorescence and transmission electron microscopy were utilized to assess the autophagic flux. Label-free quantitative phosphoproteomics was used to figure out critically altered tyrosine phosphorylation of the proteins. Tumor-bearing mice were used to validate that SH2 TrM-(Arg)9 restrained the growth of tumor cells. SH2 TrM-(Arg)9 inhibited collagen-induced autophagy via blocking the DDR1/PYK2/ERK signaling cascades. SH2 TrM-(Arg)9 improved the sensitivity of PANC-1/GEM cells to gemcitabine (GEM). Inhibition of autophagy by SH2 TrM-(Arg)9 may synergized with chemotherapy and robusted tumor suppression in pancreatic cancer xenografts. SH2 TrM-(Arg)9 could enter into PDAC cells and blockade autophagy through inhibiting DDR1/PYK2/ERK signaling and may be a new treatment strategy for targeted therapy of PDAC.
Collapse
Affiliation(s)
- Hui Xu
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
- School of Medicine, Taizhou University, 318000, Taizhou, Zhejiang, China
| | - Ming Tan
- School of Medicine, Taizhou University, 318000, Taizhou, Zhejiang, China
| | - Guo-Qing Hou
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Ya-Zhou Sang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Li Lin
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Xiao-Cai Gan
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Xuan Cao
- School of Medicine, Taizhou University, 318000, Taizhou, Zhejiang, China.
- Wenling First People's Hospital (The Affiliated Wenling Hospital of Taizhou University), School of Medicine, Taizhou University, 318000, Taizhou, Zhejiang, China.
| | - An-Dong Liu
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China.
- National Demonstration Center for Experimental Basic Medical Education, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China.
| |
Collapse
|
4
|
Hou C, Lei Y, Li N, Wei M, Wang S, Rahman SU, Bao C, Bao B, Elango J, Wu W. Collagen from Iris squid grafted with polyethylene glycol and collagen peptides promote the proliferation of fibroblast through PI3K/AKT and Ras/RAF/MAPK signaling pathways. Int J Biol Macromol 2023; 247:125772. [PMID: 37429348 DOI: 10.1016/j.ijbiomac.2023.125772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/02/2023] [Accepted: 07/07/2023] [Indexed: 07/12/2023]
Abstract
Collagens from marine sources have been used widely in food, cosmetics and tissue engineering application due to their excellent functional and biological properties. In the present study, a novel protein, collagen from iris squid skin (SSC) was characterized, grafted with polyethylene-glycol (PEG) and Acid-Green 20 (AG) and was investigated the molecular signaling pathways in L-929 fibroblast cells along with their structural peptide analogs. SDS-PAGE and IR spectrum of SSC analysis showed the typical structure of type I collagen. The fibroblast proliferation was evaluated for SSC, SSC grafted PEG (SSC-PEG) and their structural analogs including Gly-Pro-Leu-Gly-Leu-Leu (PEP1), Gly-Pro-Leu-Gly-Leu-Leu-Gly-Phe-Leu (PEP2), Gly-Pro-Leu-Gly-Leu-Leu-Gly-Phe-Leu-Gly-Pro-Leu (PEP3) and Gly-Pro-Leu-Gly-Leu-Leu-Gly-Phe-Leu-Gly-Pro-Leu-Gly-Leu-Ser (PEP4). The optimal concentration of SSC and its derivative was 0.07 μ mol/L. The fibroblast growth-promoting factors were promoted by all the treatment groups by accelerating the PI3K/AKT and Ras/RAF/MAPK signaling pathways in L-929 cells, and inhibiting the secretion of apoptotic factors. Compared to the control group, mRNA and protein expression of AKT in the PI3K/AKT and Ras in Ras/RAF/MAPK signaling pathway were accelerated significantly by PEP4, respectively, while the Bax value was significantly lower (P < 0.01). The promoting effect of PEP1, PEP2, PEP3 and PEP4 on L-929 cells was closely related to the length of the peptides. Therefore, this study disclosed that PEP1, PEP2, PEP3 and PEP4 were novel analogs that greatly promote the proliferation of L-929 cells through PI3K/AKT and Ras/RAF/MAPK signaling pathways.
Collapse
Affiliation(s)
- Chunyu Hou
- Department of Marine Bio-pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Yunjia Lei
- Department of Marine Bio-pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China.
| | - Na Li
- Department of Marine Bio-pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Mingjun Wei
- Department of Marine Bio-pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China.
| | - Shujun Wang
- Department of Marine Biopharmacology, College of Food Science and Technology, Jiangsu Ocean University, Lianyungang City 222005, Jiangsu Province, China
| | - Saeed Ur Rahman
- Oral Biology, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar 25000, Pakistan
| | - Chunling Bao
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201306, China.
| | - Bin Bao
- Zhoushan Marine Biological Engineering Co., Ltd, Zhoushan City 316104, Zhejiang Province, China.
| | - Jeevithan Elango
- Department of Marine Bio-pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; Center of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India; Department of Biomaterials Engineering, Faculty of Health Sciences, UCAM- Universidad Católica San Antonio de Murcia, Guadalupe, 30107, Murcia, Spain.
| | - Wenhui Wu
- Department of Marine Bio-pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
5
|
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
|
6
|
Wang J, Xie SA, Li N, Zhang T, Yao W, Zhao H, Pang W, Han L, Liu J, Zhou J. Matrix stiffness exacerbates the proinflammatory responses of vascular smooth muscle cell through the DDR1-DNMT1 mechanotransduction axis. Bioact Mater 2022; 17:406-424. [PMID: 35386458 PMCID: PMC8964982 DOI: 10.1016/j.bioactmat.2022.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/16/2021] [Accepted: 01/06/2022] [Indexed: 11/16/2022] Open
Abstract
Vascular smooth muscle cell (vSMC) is highly plastic as its phenotype can change in response to mechanical cues inherent to the extracellular matrix (ECM). VSMC may be activated from its quiescent contractile phenotype to a proinflammatory phenotype, whereby the cell secretes chemotactic and inflammatory cytokines, e.g. MCP1 and IL6, to functionally regulate monocyte and macrophage infiltration during the development of various vascular diseases including arteriosclerosis. Here, by culturing vSMCs on polyacrylamide (PA) substrates with variable elastic moduli, we discovered a role of discoidin domain receptor 1 (DDR1), a receptor tyrosine kinase that binds collagens, in mediating the mechanical regulation of vSMC gene expression, phenotype, and proinflammatory responses. We found that ECM stiffness induced DDR1 phosphorylation, oligomerization, and endocytosis to repress the expression of DNA methyltransferase 1 (DNMT1), very likely in a collagen-independent manner. The DDR1-to-DNMT1 signaling was sequentially mediated by the extracellular signal-regulated kinases (ERKs) and p53 pathways. ECM stiffness primed vSMC to a proinflammatory phenotype and this regulation was diminished by DDR1 inhibition. In agreement with the in vitro findings, increased DDR1 phosphorylation was observed in human arterial stiffening. DDR1 inhibition in mouse attenuated the acute injury or adenine diet-induced vascular stiffening and inflammation. Furthermore, mouse vasculature with SMC-specific deletion of Dnmt1 exhibited proinflammatory and stiffening phenotypes. Our study demonstrates a role of SMC DDR1 in perceiving the mechanical microenvironments and down-regulating expression of DNMT1 to result in vascular pathologies and has potential implications for optimization of engineering artificial vascular grafts and vascular networks. DDR1 is a mechanosensor in vSMC to perceive ECM stiffness in a collagen binding-independent way. Activation of DDR1 leads to repression of DNMT1 expression via the ERK-p53 pathway. The DDR1-DNMT1 axis mediates ECM stiffening-induced vascular inflammation.
Collapse
Affiliation(s)
- Jin Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, PR China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, PR China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, PR China
| | - Si-an Xie
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, PR China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, PR China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, PR China
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, PR China
| | - Ning Li
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), And Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, PR China
- School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, PR China
| | - Tao Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, PR China
| | - Weijuan Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, PR China
| | - Hucheng Zhao
- Institute of Biomechanics and Medical Engineering, School of Aerospace Engineering, Tsinghua University, Beijing, PR China
| | - Wei Pang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, PR China
| | - Lili Han
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, PR China
| | - Jiayu Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, PR China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, PR China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, PR China
| | - Jing Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, PR China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, PR China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, PR China
- Corresponding author. Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, PR China.
| |
Collapse
|
7
|
Itoh Y. Proteolytic modulation of tumor microenvironment signals during cancer progression. Front Oncol 2022; 12:935231. [PMID: 36132127 PMCID: PMC9483212 DOI: 10.3389/fonc.2022.935231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Under normal conditions, the cellular microenvironment is optimized for the proper functioning of the tissues and organs. Cells recognize and communicate with the surrounding cells and extracellular matrix to maintain homeostasis. When cancer arises, the cellular microenvironment is modified to optimize its malignant growth, evading the host immune system and finding ways to invade and metastasize to other organs. One means is a proteolytic modification of the microenvironment and the signaling molecules. It is now well accepted that cancer progression relies on not only the performance of cancer cells but also the surrounding microenvironment. This mini-review discusses the current understanding of the proteolytic modification of the microenvironment signals during cancer progression.
Collapse
|
8
|
Wang Y, Han B, Liu K, Wang X. Effects of DDR1 on migration and adhesion of periodontal ligament cells and the underlying mechanism. J Periodontal Res 2022; 57:568-577. [PMID: 35297053 DOI: 10.1111/jre.12986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 02/13/2022] [Accepted: 03/09/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND OBJECTIVE As one of the widely expressed cell surface receptors binding to collagen, the most abundant component of the extracellular matrix (ECM), knowledge of the expression, functions, and mechanisms underlying the role of discoidin domain receptor 1 (DDR1) in human periodontal ligament cells (hPDLCs) is incomplete. This study determined the expression of DDR1 in hPDLCs and the effect of DDR1 upon migration and adhesion to hPDLCs, as well as the related regulatory mechanisms. MATERIALS AND METHODS The expression of DDR1 and the DDR1 isoforms in hPDLCs from six donors were tested. The migratory ability (horizontal and vertical) and adhesive capacity of hPDLCs with or without specific knockdown of DDR1 were evaluated. After treatment with MEK-ERK1/2 inhibitors (PD98059 and U0126) with or without RNAi, the migratory and adhesive capacity of hPDLCs were re-tested. Western blotting was performed to verify p-MEK1/2 and p-ERK1/2, the key factors of the MEK-ERK1/2 signaling pathways. RESULTS DDR1 was detected in hPDLCs in the mRNA and protein level; DDR1b was the dominant isoform. Knockdown of DDR1 almost halved the migratory capacity and significantly downregulated the adhesive capacity of hPDLCs. The use of MEK-ERK1/2 inhibitors caused declined migratory and adhesive capacity of hPDLCs as well. After DDR1 was knocked down, the expression of p-MEK and p-ERK protein declined significantly while total MEK and ERK showed no obvious change, which means the ratio of p-MEK/MEK and p-ERK/ERK was markedly reduced. CONCLUSIONS DDR1 plays an important role in the migration and adhesion of hPDLCs and might be regulated via the MEK-ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Bing Han
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Kaining Liu
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Xiaoyan Wang
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| |
Collapse
|
9
|
The Molecular Interaction of Collagen with Cell Receptors for Biological Function. Polymers (Basel) 2022; 14:polym14050876. [PMID: 35267698 PMCID: PMC8912536 DOI: 10.3390/polym14050876] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 01/25/2023] Open
Abstract
Collagen, an extracellular protein, covers the entire human body and has several important biological functions in normal physiology. Recently, collagen from non-human sources has attracted attention for therapeutic management and biomedical applications. In this regard, both land-based animals such as cow, pig, chicken, camel, and sheep, and marine-based resources such as fish, octopus, starfish, sea-cucumber, and jellyfish are widely used for collagen extraction. The extracted collagen is transformed into collagen peptides, hydrolysates, films, hydrogels, scaffolds, sponges and 3D matrix for food and biomedical applications. In addition, many strategic ideas are continuously emerging to develop innovative advanced collagen biomaterials. For this purpose, it is important to understand the fundamental perception of how collagen communicates with receptors of biological cells to trigger cell signaling pathways. Therefore, this review discloses the molecular interaction of collagen with cell receptor molecules to carry out cellular signaling in biological pathways. By understanding the actual mechanism, this review opens up several new concepts to carry out next level research in collagen biomaterials.
Collapse
|
10
|
Sannomiya Y, Kaseda S, Kamura M, Yamamoto H, Yamada H, Inamoto M, Kuwazuru J, Niino S, Shuto T, Suico MA, Kai H. The role of discoidin domain receptor 2 in the renal dysfunction of alport syndrome mouse model. Ren Fail 2021; 43:510-519. [PMID: 33706638 PMCID: PMC7971217 DOI: 10.1080/0886022x.2021.1896548] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/09/2021] [Accepted: 02/19/2021] [Indexed: 02/08/2023] Open
Abstract
Alport syndrome (AS) is a hereditary glomerular nephritis caused by mutation in one of the type IV collagen genes α3/α4/α5 that encode the heterotrimer COL4A3/4/5. Failure to form a heterotrimer due to mutation leads to the dysfunction of the glomerular basement membrane, and end-stage renal disease. Previous reports have suggested the involvement of the receptor tyrosine kinase discoidin domain receptor (DDR) 1 in the progression of AS pathology. However, due to the similarity between DDR1 and DDR2, the role of DDR2 in AS pathology is unclear. Here, we investigated the involvement of DDR2 in AS using the X-linked AS mouse model. Mice were treated subcutaneously with saline or antisense oligonucleotide (ASO; 5 mg/kg or 15 mg/kg per week) for 8 weeks. Renal function parameters and renal histology were analyzed, and the gene expressions of inflammatory cytokines were determined in renal tissues. The expression level of DDR2 was highly elevated in kidney tissues of AS mice. Knockdown of Ddr2 using Ddr2-specific ASO decreased the Ddr2 expression. However, the DDR2 ASO treatment did not improve the proteinuria or decrease the BUN level. DDR2 ASO also did not significantly ameliorate the renal injury, inflammation and fibrosis in AS mice. These results showed that Ddr2 knockdown by ASO had no notable effect on the progression of AS indicating that DDR2 may not be critically involved in AS pathology. This finding may provide useful information and further understanding of the role of DDRs in AS.
Collapse
Affiliation(s)
- Yuya Sannomiya
- Department of Molecular Medicine Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
| | - Shota Kaseda
- Department of Molecular Medicine Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
- Program for Leading Graduate Schools “HIGO (Health life science: Interdisciplinary and Glocal Oriented) Program”, Kumamoto University, Kumamoto, Japan
| | - Misato Kamura
- Department of Molecular Medicine Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
- Program for Leading Graduate Schools “HIGO (Health life science: Interdisciplinary and Glocal Oriented) Program”, Kumamoto University, Kumamoto, Japan
| | | | | | | | - Jun Kuwazuru
- Department of Molecular Medicine Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
| | - Saki Niino
- Department of Molecular Medicine Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
| | - Tsuyoshi Shuto
- Department of Molecular Medicine Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
- Global Center for Natural Resources Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Mary Ann Suico
- Department of Molecular Medicine Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
- Global Center for Natural Resources Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hirofumi Kai
- Department of Molecular Medicine Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
- Program for Leading Graduate Schools “HIGO (Health life science: Interdisciplinary and Glocal Oriented) Program”, Kumamoto University, Kumamoto, Japan
- Global Center for Natural Resources Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
11
|
Itoh Y. Modulation of Microenvironment Signals by Proteolytic Shedding of Cell Surface Extracellular Matrix Receptors. Front Cell Dev Biol 2021; 9:736735. [PMID: 34796172 PMCID: PMC8593224 DOI: 10.3389/fcell.2021.736735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/20/2021] [Indexed: 01/02/2023] Open
Abstract
Multicellular organisms are composed of cells and extracellular matrix (ECM). ECM is a network of multidomain macromolecules that fills gaps between cells. It acts as a glue to connect cells, provides scaffolding for migrating cells, and pools cytokines and growth factors. ECM also directly sends signals to the cells through ECM receptors, providing survival signals and migration cues. Altogether, ECM provides a correct microenvironment for the cells to function in the tissue. Although ECM acts as a signaling molecule, they are insoluble solid molecules, unlike soluble receptor ligands such as cytokines and growth factors. Upon cell binding to the ECM through ECM receptors and signals transmitted, cells then need to have a mechanism to release from ECM to prevent prolonged signals, which may be tumorigenic, and migrate on ECM. One effective means to release the cells from ECM is to cleave the ECM receptors by proteinases. In this mini-review, current knowledge of ECM receptor shedding will be discussed.
Collapse
Affiliation(s)
- Yoshifumi Itoh
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
12
|
Ferrao Blanco MN, Domenech Garcia H, Legeai-Mallet L, van Osch GJVM. Tyrosine kinases regulate chondrocyte hypertrophy: promising drug targets for Osteoarthritis. Osteoarthritis Cartilage 2021; 29:1389-1398. [PMID: 34284112 DOI: 10.1016/j.joca.2021.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/24/2021] [Accepted: 07/08/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is a major health problem worldwide that affects the joints and causes severe disability. It is characterized by pain and low-grade inflammation. However, the exact pathogenesis remains unknown and the therapeutic options are limited. In OA articular chondrocytes undergo a phenotypic transition becoming hypertrophic, which leads to cartilage damage, aggravating the disease. Therefore, a therapeutic agent inhibiting hypertrophy would be a promising disease-modifying drug. The therapeutic use of tyrosine kinase inhibitors has been mainly focused on oncology, but the Food and Drug Administration (FDA) approval of the Janus kinase inhibitor Tofacitinib in Rheumatoid Arthritis has broadened the applicability of these compounds to other diseases. Interestingly, tyrosine kinases have been associated with chondrocyte hypertrophy. In this review, we discuss the experimental evidence that implicates specific tyrosine kinases in signaling pathways promoting chondrocyte hypertrophy, highlighting their potential as therapeutic targets for OA.
Collapse
Affiliation(s)
- M N Ferrao Blanco
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - H Domenech Garcia
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - L Legeai-Mallet
- Université de Paris, INSERM U1163, Institut Imagine, Paris, France.
| | - G J V M van Osch
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Biomechanical Engineering, Delft University of Technology, Delft, the Netherlands.
| |
Collapse
|
13
|
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: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
14
|
Orgel JPRO, Madhurapantula RS. A structural prospective for collagen receptors such as DDR and their binding of the collagen fibril. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:118478. [PMID: 31004686 DOI: 10.1016/j.bbamcr.2019.04.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 04/11/2019] [Accepted: 04/12/2019] [Indexed: 12/13/2022]
Abstract
The structure of the collagen fibril surface directly effects and possibly assists the management of collagen receptor interactions. An important class of collagen receptors, the receptor tyrosine kinases of the Discoidin Domain Receptor family (DDR1 and DDR2), are differentially activated by specific collagen types and play important roles in cell adhesion, migration, proliferation, and matrix remodeling. This review discusses their structure and function as it pertains directly to the fibrillar collagen structure with which they interact far more readily than they do with isolated molecular collagen. This prospective provides further insight into the mechanisms of activation and rational cellular control of this important class of receptors while also providing a comparison of DDR-collagen interactions with other receptors such as integrin and GPVI. When improperly regulated, DDR activation can lead to abnormal cellular proliferation activities such as in cancer. Hence how and when the DDRs associate with the major basis of mammalian tissue infrastructure, fibrillar collagen, should be of keen interest.
Collapse
Affiliation(s)
- Joseph P R O Orgel
- Departments of Biology and Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA.
| | - Rama S Madhurapantula
- Departments of Biology and Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| |
Collapse
|
15
|
Moll S, Desmoulière A, Moeller MJ, Pache JC, Badi L, Arcadu F, Richter H, Satz A, Uhles S, Cavalli A, Drawnel F, Scapozza L, Prunotto M. DDR1 role in fibrosis and its pharmacological targeting. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:118474. [PMID: 30954571 DOI: 10.1016/j.bbamcr.2019.04.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 01/28/2023]
Abstract
Discoidin domain receptor1 (DDR1) is a collagen activated receptor tyrosine kinase and an attractive anti-fibrotic target. Its expression is mainly limited to epithelial cells located in several organs including skin, kidney, liver and lung. DDR1's biology is elusive, with unknown downstream activation pathways; however, it may act as a mediator of the stromal-epithelial interaction, potentially controlling the activation state of the resident quiescent fibroblasts. Increased expression of DDR1 has been documented in several types of cancer and fibrotic conditions including skin hypertrophic scars, idiopathic pulmonary fibrosis, cirrhotic liver and renal fibrosis. The present review article focuses on: a) detailing the evidence for a role of DDR1 as an anti-fibrotic target in different organs, b) clarifying DDR1 tissue distribution in healthy and diseased tissues as well as c) exploring DDR1 protective mode of action based on literature evidence and co-authors experience; d) detailing pharmacological efforts attempted to drug this subtle anti-fibrotic target to date.
Collapse
Affiliation(s)
- Solange Moll
- Department of Pathology, University Hospital of Geneva, Switzerland; Department of Pathology, Lausanne University Hospital, Switzerland
| | - Alexis Desmoulière
- Department of Physiology, Faculty of Pharmacy, University of Limoges, Limoges, France
| | - Marcus J Moeller
- Department of Nephrology and Clinical Immunology, RWTH University Hospital, Aachen, Germany
| | | | - Laura Badi
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Switzerland
| | - Filippo Arcadu
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Switzerland
| | - Hans Richter
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Switzerland
| | - Alexander Satz
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Switzerland
| | - Sabine Uhles
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Switzerland
| | - Andrea Cavalli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, CH-6500, Bellinzona, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Faye Drawnel
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Switzerland
| | - Leonardo Scapozza
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Marco Prunotto
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Switzerland; School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
16
|
Henriet E, Sala M, Abou Hammoud A, Tuariihionoa A, Di Martino J, Ros M, Saltel F. Multitasking discoidin domain receptors are involved in several and specific hallmarks of cancer. Cell Adh Migr 2018; 12:363-377. [PMID: 29701112 PMCID: PMC6411096 DOI: 10.1080/19336918.2018.1465156] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/28/2018] [Accepted: 04/03/2018] [Indexed: 12/11/2022] Open
Abstract
Discoidin domain receptors, DDR1 and DDR2, are two members of collagen receptor family that belong to tyrosine kinase receptor subgroup. Unlike other matrix receptor-like integrins, these collagen receptors have not been extensively studied. However, more and more studies are focusing on their involvement in cancer. These two receptors are present in several subcellular localizations such as intercellular junction or along type I collagen fibers. Consequently, they are involved in multiple cellular functions, for instance, cell cohesion, proliferation, adhesion, migration and invasion. Furthermore, various signaling pathways are associated with these multiple functions. In this review, we highlight and characterize hallmarks of cancer in which DDRs play crucial roles. We discuss recent data from studies that demonstrate the involvement of DDRs in tumor proliferation, cancer mutations, drug resistance, inflammation, neo-angiogenesis and metastasis. DDRs could be potential targets in cancer and we conclude this review by discussing the different ways to inhibits them.
Collapse
Affiliation(s)
- Elodie Henriet
- INSERM, UMR1053, BaRITOn Bordeaux Research in Translational Oncology,Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| | - Margaux Sala
- INSERM, UMR1053, BaRITOn Bordeaux Research in Translational Oncology,Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| | - Aya Abou Hammoud
- INSERM, UMR1053, BaRITOn Bordeaux Research in Translational Oncology,Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| | - Adjanie Tuariihionoa
- INSERM, UMR1053, BaRITOn Bordeaux Research in Translational Oncology,Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| | - Julie Di Martino
- INSERM, UMR1053, BaRITOn Bordeaux Research in Translational Oncology,Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| | - Manon Ros
- INSERM, UMR1053, BaRITOn Bordeaux Research in Translational Oncology,Bordeaux, France
- Université de Bordeaux, Bordeaux, France
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore
| | - Frédéric Saltel
- INSERM, UMR1053, BaRITOn Bordeaux Research in Translational Oncology,Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| |
Collapse
|
17
|
Itoh Y. Discoidin domain receptors: Microenvironment sensors that promote cellular migration and invasion. Cell Adh Migr 2018; 12:378-385. [PMID: 29671358 PMCID: PMC6363040 DOI: 10.1080/19336918.2018.1460011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Extracellular matrix (ECM) provides cells scaffolding for cell migration and microenvironment for various cellular functions. Collagens are major ECM components in tissue and discoidin domain receptors (DDRs) are receptor tyrosine kinases (RTK) that recognise fibrillar collagens. Unlike other RTK, their ligands are solid ECM the that are abundantly present in the pericellular environment in various tissue, and thus its activation and regulations are unique amongst RTK family. It is emerging that DDRs may be the sensors that monitor and detects changes in ECM microenvironment and determines the cellular fates upon tissue injuries. In this mini-review, recent findings on the role of DDRs as microenvironment sensor and their roles in cell migration and invasion are discussed.
Collapse
Affiliation(s)
- Yoshifumi Itoh
- a Kennedy Institute of Rheumatology, University of Oxford , Roosevelt Drive, Headington , Oxford , UK
| |
Collapse
|
18
|
Chen C, Deng J, Yu X, Wu F, Men K, Yang Q, Zhu Y, Liu X, Jiang Q. Identification of novel inhibitors of DDR1 against idiopathic pulmonary fibrosis by integrative transcriptome meta-analysis, computational and experimental screening. MOLECULAR BIOSYSTEMS 2017; 12:1540-51. [PMID: 26956955 DOI: 10.1039/c5mb00911a] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a kind of a chronic and fatal lung disease leading to progressive lung function decline. Although several RNA microarray studies on IPF patients have been reported, their results were merely specific to each study with distinct platforms or sample types. In the current study, an integrative transcriptome meta-analysis of IPF was performed to explore regulated pathways, based on four independent expression profiling microarrays of IPF datasets, including 73 samples from IPF tissues or lung fibroblast cells. The results suggested the discoidin domain receptor 1 (DDR1) and downstream c-Jun N-terminal kinases (JNK) pathway may play important roles in the progression of IPF. To our knowledge, discoidin domain receptor 1 (DDR1) is a kind of receptor tyrosine kinase (RTK) with a unique ability to bind both fibrillar and non-fibrillar collagens. Based on the crystallographic structures of DDR1, the combination of molecular dynamics simulation and a hybrid protocol of a virtual screening method, comprised of PBVS (multicomplex-pharmacophore based virtual screening) and DBVS (docking based virtual screening) methods were used for retrieving novel DDR1 inhibitors from the SPECS database. Twelve hit compounds were selected from the hit compounds and shifted to experimental validations, and the most potent compound was evaluated for its anti-IPF capacity on murine IPF models. Thus, these results may provide valuable information for further discovery of potential lead compounds for IPF therapy.
Collapse
Affiliation(s)
- Can Chen
- School of Pharmacy and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, 610050, P. R. China.
| | - Jingjing Deng
- School of Pharmacy and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, 610050, P. R. China.
| | - Xiaoping Yu
- School of Pharmacy and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, 610050, P. R. China. and Department of Public Health, Chengdu Medical College, Chengdu, 610050, P. R. China
| | - Fengbo Wu
- State Key laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Ke Men
- State Key laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Qian Yang
- School of Pharmacy and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, 610050, P. R. China.
| | - Yanfeng Zhu
- School of Pharmacy and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, 610050, P. R. China. and Department of Public Health, Chengdu Medical College, Chengdu, 610050, P. R. China
| | - Xiaogang Liu
- Department of Gastroenterology, Hospital of the University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, 610065, P. R. China
| | - Qinglin Jiang
- School of Pharmacy and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, 610050, P. R. China. and State Key laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
19
|
Extracellular matrix component signaling in cancer. Adv Drug Deliv Rev 2016; 97:28-40. [PMID: 26519775 DOI: 10.1016/j.addr.2015.10.013] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 12/12/2022]
Abstract
Cell responses to the extracellular matrix depend on specific signaling events. These are important from early development, through differentiation and tissue homeostasis, immune surveillance, and disease pathogenesis. Signaling not only regulates cell adhesion cytoskeletal organization and motility but also provides survival and proliferation cues. The major classes of cell surface receptors for matrix macromolecules are the integrins, discoidin domain receptors, and transmembrane proteoglycans such as syndecans and CD44. Cells respond not only to specific ligands, such as collagen, fibronectin, or basement membrane glycoproteins, but also in terms of matrix rigidity. This can regulate the release and subsequent biological activity of matrix-bound growth factors, for example, transforming growth factor-β. In the environment of tumors, there may be changes in cell populations and their receptor profiles as well as matrix constitution and protein cross-linking. Here we summarize roles of the three major matrix receptor types, with emphasis on how they function in tumor progression.
Collapse
|
20
|
Krohn JB, Hutcheson JD, Martínez-Martínez E, Irvin WS, Bouten CVC, Bertazzo S, Bendeck MP, Aikawa E. Discoidin Domain Receptor-1 Regulates Calcific Extracellular Vesicle Release in Vascular Smooth Muscle Cell Fibrocalcific Response via Transforming Growth Factor-β Signaling. Arterioscler Thromb Vasc Biol 2016; 36:525-33. [PMID: 26800565 DOI: 10.1161/atvbaha.115.307009] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 01/06/2015] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Collagen accumulation and calcification are major determinants of atherosclerotic plaque stability. Extracellular vesicle (EV)-derived microcalcifications in the collagen-poor fibrous cap may promote plaque rupture. In this study, we hypothesize that the collagen receptor discoidin domain receptor-1 (DDR-1) regulates collagen deposition and release of calcifying EVs by vascular smooth muscle cells (SMCs) through the transforming growth factor-β (TGF-β) pathway. APPROACH AND RESULTS SMCs from the carotid arteries of DDR-1(-/-) mice and wild-type littermates (n=5-10 per group) were cultured in normal or calcifying media. At days 14 and 21, SMCs were harvested and EVs isolated for analysis. Compared with wild-type, DDR-1(-/-) SMCs exhibited a 4-fold increase in EV release (P<0.001) with concomitantly elevated alkaline phosphatase activity (P<0.0001) as a hallmark of EV calcifying potential. The DDR-1(-/-) phenotype was characterized by increased mineralization (Alizarin Red S and Osteosense, P<0.001 and P=0.002, respectively) and amorphous collagen deposition (P<0.001). We further identified a novel link between DDR-1 and the TGF-β pathway previously implicated in both fibrotic and calcific responses. An increase in TGF-β1 release by DDR-1(-/-) SMCs in calcifying media (P<0.001) stimulated p38 phosphorylation (P=0.02) and suppressed activation of Smad3. Inhibition of either TGF-β receptor-I or phospho-p38 reversed the fibrocalcific DDR-1(-/-) phenotype, corroborating a causal relationship between DDR-1 and TGF-β in EV-mediated vascular calcification. CONCLUSIONS DDR-1 interacts with the TGF-β pathway to restrict calcifying EV-mediated mineralization and fibrosis by SMCs. We therefore establish a novel mechanism of cell-matrix homeostasis in atherosclerotic plaque formation.
Collapse
Affiliation(s)
- Jona B Krohn
- From the Department of Medicine, Cardiovascular Division, Center for Excellence in Vascular Biology (J.B.K., E.M.-M., W.S.I., E.A.) and Center for Interdisciplinary Cardiovascular Sciences (J.D.H., E.A.), Harvard Medical School, Boston, MA; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands (C.V.C.B.); Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom (S.B.); and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada (M.P.B.)
| | - Joshua D Hutcheson
- From the Department of Medicine, Cardiovascular Division, Center for Excellence in Vascular Biology (J.B.K., E.M.-M., W.S.I., E.A.) and Center for Interdisciplinary Cardiovascular Sciences (J.D.H., E.A.), Harvard Medical School, Boston, MA; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands (C.V.C.B.); Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom (S.B.); and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada (M.P.B.)
| | - Eduardo Martínez-Martínez
- From the Department of Medicine, Cardiovascular Division, Center for Excellence in Vascular Biology (J.B.K., E.M.-M., W.S.I., E.A.) and Center for Interdisciplinary Cardiovascular Sciences (J.D.H., E.A.), Harvard Medical School, Boston, MA; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands (C.V.C.B.); Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom (S.B.); and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada (M.P.B.)
| | - Whitney S Irvin
- From the Department of Medicine, Cardiovascular Division, Center for Excellence in Vascular Biology (J.B.K., E.M.-M., W.S.I., E.A.) and Center for Interdisciplinary Cardiovascular Sciences (J.D.H., E.A.), Harvard Medical School, Boston, MA; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands (C.V.C.B.); Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom (S.B.); and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada (M.P.B.)
| | - Carlijn V C Bouten
- From the Department of Medicine, Cardiovascular Division, Center for Excellence in Vascular Biology (J.B.K., E.M.-M., W.S.I., E.A.) and Center for Interdisciplinary Cardiovascular Sciences (J.D.H., E.A.), Harvard Medical School, Boston, MA; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands (C.V.C.B.); Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom (S.B.); and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada (M.P.B.)
| | - Sergio Bertazzo
- From the Department of Medicine, Cardiovascular Division, Center for Excellence in Vascular Biology (J.B.K., E.M.-M., W.S.I., E.A.) and Center for Interdisciplinary Cardiovascular Sciences (J.D.H., E.A.), Harvard Medical School, Boston, MA; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands (C.V.C.B.); Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom (S.B.); and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada (M.P.B.)
| | - Michelle P Bendeck
- From the Department of Medicine, Cardiovascular Division, Center for Excellence in Vascular Biology (J.B.K., E.M.-M., W.S.I., E.A.) and Center for Interdisciplinary Cardiovascular Sciences (J.D.H., E.A.), Harvard Medical School, Boston, MA; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands (C.V.C.B.); Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom (S.B.); and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada (M.P.B.)
| | - Elena Aikawa
- From the Department of Medicine, Cardiovascular Division, Center for Excellence in Vascular Biology (J.B.K., E.M.-M., W.S.I., E.A.) and Center for Interdisciplinary Cardiovascular Sciences (J.D.H., E.A.), Harvard Medical School, Boston, MA; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands (C.V.C.B.); Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom (S.B.); and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada (M.P.B.).
| |
Collapse
|
21
|
Li W, Guo A, Wang L, Kong Q, Wang R, Han L, Zhao C. Expression of peptide fragments from proADM and involvement of mitogen-activated protein kinase signaling pathways in pulmonary remodeling induced by high pulmonary blood flow. Congenit Anom (Kyoto) 2016; 56:28-34. [PMID: 25990643 DOI: 10.1111/cga.12114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 05/09/2015] [Indexed: 01/02/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by progressive pulmonary arterial remodeling and right ventricular failure. Despite recent advances in pathophysiological mechanism exploration and new therapeutic approaches, PAH remains a challenging condition. In this study, we investigated the roles of the peptide fragments from proadrenomedullin (proADM) such as adrenomedullin (ADM), adrenotensin (ADT), and proadrenomedullin N-terminal 20 peptide (PAMP) during pulmonary remodeling caused by high pulmonary blood flow, and probed the possible involvement of mitogen-activated protein kinase (MAPK) signal transduction pathways. Sixteen rat models of PAH were artificially established by surgically connecting the left common carotid artery to the external jugular vein. We subcutaneously injected an extracellular signal-regulated protein kinase (ERK1/2) inhibitor, PD98059, in eight rats, treated another eight rats with an equal volume of saline. Eight rats without connections served as the control group. We observed that mRNA expression levels of ADM, stress-activated protein kinase (SAPK), and ERK1/2 were significantly elevated in the shunted rats; furthermore, ERK1/2 levels were significantly inhibited by PD98059. Protein levels of ADM, PAMP, p-SAPK, and p-ERK1/2 were significantly higher ADT was lower, and p-p38 remained unchanged in the rat models compared with the controls. However, the protein expression of both ADM and p-ERK1/2 was significantly inhibited by PD98059. Our results suggest that levels of ADM, ADT, and PAMP respond to pulmonary remodeling, and that activation of the SAPK and ERK1/2 signaling pathways is involved in pulmonary hypertension and artery remodeling caused by high pulmonary blood flow.
Collapse
Affiliation(s)
- Wei Li
- Biomedical Engineering Institute, School of Control Science and Engineering, Shandong University, Jinan, China
| | - Aili Guo
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, China
| | - Lijuan Wang
- Beijing Children's Hospital Affiliated to Capital Medical University, Beijing, China
| | - Qingyu Kong
- Biomedical Engineering Institute, School of Control Science and Engineering, Shandong University, Jinan, China
| | - Rong Wang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China
| | - Li Han
- Department of Ophthalmology, Yidu Central Hospital of Weifang, Qingzhou, China
| | - Cuifen Zhao
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
22
|
Urotensin II contributes to collagen synthesis and up-regulates Egr-1 expression in cultured pulmonary arterial smooth muscle cells through the ERK1/2 pathway. Biochem Biophys Res Commun 2015; 467:1076-82. [DOI: 10.1016/j.bbrc.2015.09.148] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 09/27/2015] [Indexed: 12/18/2022]
|
23
|
High expression of DDR1 is associated with the poor prognosis in Chinese patients with pancreatic ductal adenocarcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:88. [PMID: 26297342 PMCID: PMC4546266 DOI: 10.1186/s13046-015-0202-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 08/06/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Discoidin domain receptors 1 (DDR1), a subtype of DDRs, has been reported as a critical modulator of cellular morphogenesis, differentiation, migration and invasion. METHODS AND RESULTS In this study, we investigated the expression of DDR1 and its clinical association in Chinese patients with pancreatic ductal adenocarcinoma (PDAC). Across a cohort of 30 patients, we examined DDR1 expression in paired PDAC and corresponding adjacent non-tumor tissues by real-time quantitative PCR (RT-qPCR), or western blotting. DDR1 expression is significantly higher in PDAC, as compared to normal adjacent tissue, confirming results from the Oncomine databases. We validated DDR1 expression by immunohistochemistry across a non-overlapping cohort of 205 PDAC specimens. Kaplan-Meier survival curves indicate that increased expression of DDR1 is associated with a poor prognosis in PDAC patients (P = 0.013). Multivariate Cox regression analysis identified DDR1 expression, age, N classification and liver metastasis as independent prognostic factors in PDAC. CONCLUSIONS This study demonstrated that DDR1 can well serve as a novel prognostic biomarker in PDAC.
Collapse
|
24
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Vascular smooth muscle cell in atherosclerosis. Acta Physiol (Oxf) 2015; 214:33-50. [PMID: 25677529 DOI: 10.1111/apha.12466] [Citation(s) in RCA: 294] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/02/2015] [Accepted: 02/09/2015] [Indexed: 12/30/2022]
Abstract
Vascular smooth muscle cells (VSMCs) exhibit phenotypic and functional plasticity in order to respond to vascular injury. In case of the vessel damage, VSMCs are able to switch from the quiescent 'contractile' phenotype to the 'proinflammatory' phenotype. This change is accompanied by decrease in expression of smooth muscle (SM)-specific markers responsible for SM contraction and production of proinflammatory mediators that modulate induction of proliferation and chemotaxis. Indeed, activated VSMCs could efficiently proliferate and migrate contributing to the vascular wall repair. However, in chronic inflammation that occurs in atherosclerosis, arterial VSMCs become aberrantly regulated and this leads to increased VSMC dedifferentiation and extracellular matrix formation in plaque areas. Proatherosclerotic switch in VSMC phenotype is a complex and multistep mechanism that may be induced by a variety of proinflammatory stimuli and hemodynamic alterations. Disturbances in hemodynamic forces could initiate the proinflammatory switch in VSMC phenotype even in pre-clinical stages of atherosclerosis. Proinflammatory signals play a crucial role in further dedifferentiation of VSMCs in affected vessels and propagation of pathological vascular remodelling.
Collapse
Affiliation(s)
- D. A. Chistiakov
- Research Center for Children's Health; Moscow Russia
- The Mount Sinai Community Clinical Oncology Program; Mount Sinai Comprehensive Cancer Center; Mount Sinai Medical Center; Miami Beach FL USA
| | - A. N. Orekhov
- Institute for Atherosclerosis; Skolkovo Innovative Center; Moscow Russia
- Laboratory of Angiopathology; Institute of General Pathology and Pathophysiology; Russian Academy of Sciences; Moscow Russia
- Department of Biophysics; Biological Faculty; Moscow State University; Moscow Russia
| | - Y. V. Bobryshev
- Institute for Atherosclerosis; Skolkovo Innovative Center; Moscow Russia
- Faculty of Medicine; School of Medical Sciences; University of New South Wales; Kensington Sydney NSW Australia
- School of Medicine; University of Western Sydney; Campbelltown NSW Australia
| |
Collapse
|
25
|
Elliott KJ, Eguchi S. Phosphorylation Regulation by Kinases and Phosphatases in Atherosclerosis. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
26
|
Discoidin domain receptors (DDRs): Potential implications in atherosclerosis. Eur J Pharmacol 2015; 751:28-33. [DOI: 10.1016/j.ejphar.2015.01.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 01/19/2015] [Accepted: 01/21/2015] [Indexed: 01/15/2023]
|
27
|
Collagen inhibitory peptide R1R2 mediates vascular remodeling by decreasing inflammation and smooth muscle cell activation. PLoS One 2015; 10:e0117356. [PMID: 25675397 PMCID: PMC4326127 DOI: 10.1371/journal.pone.0117356] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Accepted: 12/22/2014] [Indexed: 11/30/2022] Open
Abstract
The extracellular matrix (ECM) is a major constituent of the vessel wall. In addition to providing a structural scaffold, the ECM controls numerous cellular functions in both physiologic and pathologic settings. Vascular remodeling occurs after injury and is characterized by endothelial cell activation, inflammatory cell infiltration, phenotypic modulation of smooth muscle cells (SMCs), and augmented deposition of collagen-rich ECM. R1R2, a peptide derived from the bacterial adhesin SFS, with sequence homology to collagen, is known to inhibit collagen type I deposition in vitro by inhibiting the binding of fibronectin to collagen. However, the inhibitory effects of R1R2 during vascular remodeling have not been explored. We periadventitially delivered R1R2 to carotid arteries using pluronic gel in a vascular remodeling mouse model induced by blood flow cessation, and evaluated its effects on intima-media thickening, ECM deposition, SMC activation, and inflammatory cell infiltration. Morphometric analysis demonstrated that R1R2 reduced intima-media thickening compared to the control groups. R1R2 treatment also decreased collagen type I deposition in the vessel wall, and maintained SMC in the contractile phenotype. Interestingly, R1R2 dramatically reduced inflammatory cell infiltration into the vessel by ∼78%. This decrease was accompanied by decreased VCAM-1 and ICAM-1 expression. Our in vitro studies revealed that R1R2 attenuated SMC proliferation and migration, and also decreased monocyte adhesion and transendothelial migration through endothelial cells. Together, these data suggest that R1R2 attenuates vascular remodeling responses by decreasing inflammation and by modulating SMC proliferation and migration, and suggest that the R1R2 peptide may have therapeutic potential in treating occlusive vascular diseases.
Collapse
|
28
|
Kothiwale S, Borza CM, Lowe EW, Pozzi A, Meiler J. Discoidin domain receptor 1 (DDR1) kinase as target for structure-based drug discovery. Drug Discov Today 2015; 20:255-61. [PMID: 25284748 PMCID: PMC4336622 DOI: 10.1016/j.drudis.2014.09.025] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 08/08/2014] [Accepted: 09/26/2014] [Indexed: 11/18/2022]
Abstract
Discoidin domain receptor (DDR) 1 and 2 are transmembrane receptors that belong to the family of receptor tyrosine kinases (RTK). Upon collagen binding, DDRs transduce cellular signaling involved in various cell functions, including cell adhesion, proliferation, differentiation, migration, and matrix homeostasis. Altered DDR function resulting from either mutations or overexpression has been implicated in several types of disease, including atherosclerosis, inflammation, cancer, and tissue fibrosis. Several established inhibitors, such as imatinib, dasatinib, and nilotinib, originally developed as Abelson murine leukemia (Abl) kinase inhibitors, have been found to inhibit DDR kinase activity. As we review here, recent discoveries of novel inhibitors and their co-crystal structure with the DDR1 kinase domain have made structure-based drug discovery for DDR1 amenable.
Collapse
Affiliation(s)
| | - Corina M Borza
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN 37232, USA
| | - Edward W Lowe
- Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Ambra Pozzi
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN 37232, USA; Department of Medicine, Veterans Affairs Hospital, Nashville, TN 37232, USA
| | - Jens Meiler
- Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
29
|
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
|
30
|
Shitomi Y, Thøgersen IB, Ito N, Leitinger B, Enghild JJ, Itoh Y. ADAM10 controls collagen signaling and cell migration on collagen by shedding the ectodomain of discoidin domain receptor 1 (DDR1). Mol Biol Cell 2014; 26:659-73. [PMID: 25540428 PMCID: PMC4325837 DOI: 10.1091/mbc.e14-10-1463] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Collagen receptor DDR1 is shed upon collagen binding by ADAM10 metalloproteinase. This shedding controls the half-life of DDR1 signaling and cell migration on the collagen matrix. This event may be a part of a regulatory mechanism of microenvironment signaling. Discoidin domain receptor 1 (DDR1) is a receptor tyrosine kinase that binds and transmits signals from various collagens in epithelial cells. However, how DDR1–dependent signaling is regulated has not been understood. Here we report that collagen binding induces ADAM10-dependent ectodomain shedding of DDR1. DDR1 shedding is not a result of an activation of its signaling pathway, since DDR1 mutants defective in signaling were shed in an efficient manner. DDR1 and ADAM10 were found to be in a complex on the cell surface, but shedding did not occur unless collagen bound to DDR1. Using a shedding-resistant DDR1 mutant, we found that ADAM10-dependent DDR1 shedding regulates the half-life of collagen-induced phosphorylation of the receptor. Our data also revealed that ADAM10 plays an important role in regulating DDR1-mediated cell adhesion to achieve efficient cell migration on collagen matrices.
Collapse
Affiliation(s)
- Yasuyuki Shitomi
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, United Kingdom
| | - Ida B Thøgersen
- Department of Molecular Biology and Genetics, University of Aarhus, DK-8000 Aarhus C, Denmark
| | - Noriko Ito
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, United Kingdom
| | - Birgit Leitinger
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, United Kingdom
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, University of Aarhus, DK-8000 Aarhus C, Denmark
| | - Yoshifumi Itoh
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, United Kingdom
| |
Collapse
|
31
|
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: 259] [Impact Index Per Article: 23.5] [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
|
32
|
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: 11.1] [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
|
33
|
Li W, Kong QY, Zhao CF, Zhao F, Li FH, Xia W, Wang R, Hu YM, Hua M. Adrenomedullin and adrenotensin regulate collagen synthesis and proliferation in pulmonary arterial smooth muscle cells. Braz J Med Biol Res 2013; 46:1047-1055. [PMID: 24345914 PMCID: PMC3935277 DOI: 10.1590/1414-431x20132882] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 07/29/2013] [Indexed: 11/22/2022] Open
Abstract
To understand the pathophysiological mechanisms of pulmonary arterial smooth muscle cell (PASMC) proliferation and extracellular-matrix accumulation in the development of pulmonary hypertension and remodeling, this study determined the effects of different doses of adrenomedullin (ADM) and adrenotensin (ADT) on PASMC proliferation and collagen synthesis. The objective was to investigate whether extracellular signal-regulated kinase (ERK1/2) signaling was involved in ADM- and ADT-stimulated proliferation of PASMCs in 4-week-old male Wistar rats (body weight: 100-150 g, n=10). The proliferation of PASMCs was examined by 5-bromo-2-deoxyuridine incorporation. A cell growth curve was generated by the Cell Counting Kit-8 method. Expression of collagen I, collagen III, and phosphorylated ERK1/2 (p-ERK1/2) was evaluated by immunofluorescence. The effects of different concentrations of ADM and ADT on collagen I, collagen III, and p-ERK1/2 protein expression were determined by immunoblotting. We also investigated the effect of PD98059 inhibition on the expression of p-ERK1/2 protein by immunoblotting. ADM dose-dependently decreased cell proliferation, whereas ADT dose-dependently increased it; and ADM and ADT inhibited each other with respect to their effects on the proliferation of PASMCs. Consistent with these results, the expression of collagen I, collagen III, and p-ERK1/2 in rat PASMCs decreased after exposure to ADM but was upregulated after exposure to ADT. PD98059 significantly inhibited the downregulation by ADM and the upregulation by ADT of p-ERK1/2 expression. We conclude that ADM inhibited, and ADT stimulated, ERK1/2 signaling in rat PASMCs to regulate cell proliferation and collagen expression.
Collapse
Affiliation(s)
- W Li
- Shandong University, Biomedical Engineering Institute, School of Control Science and Engineering, JinanShandong, China
| | - Q Y Kong
- Shandong University, Qilu Hospital, Department of Pediatrics, JinanShandong, China
| | - C F Zhao
- Shandong University, Qilu Hospital, Department of Pediatrics, JinanShandong, China
| | - F Zhao
- Weill Medical College of Cornell University, Department of Medicine, New YorkNY, USA
| | - F H Li
- Shandong University, Qilu Hospital, Department of Pediatrics, JinanShandong, China
| | - W Xia
- Shandong University, Qilu Hospital, Department of Pediatrics, JinanShandong, China
| | - R Wang
- Shandong University, Qilu Hospital, Key Laboratory of Cardiovascular Remodeling and Function Research, JinanShandong, China
| | - Y M Hu
- Shandong University, Biomedical Engineering Institute, School of Control Science and Engineering, JinanShandong, China
| | - M Hua
- Shandong Institute of Scientific and Technical Information, JinanShandong, China
| |
Collapse
|
34
|
Koohestani F, Braundmeier AG, Mahdian A, Seo J, Bi J, Nowak RA. Extracellular matrix collagen alters cell proliferation and cell cycle progression of human uterine leiomyoma smooth muscle cells. PLoS One 2013; 8:e75844. [PMID: 24040420 PMCID: PMC3770620 DOI: 10.1371/journal.pone.0075844] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 08/22/2013] [Indexed: 12/25/2022] Open
Abstract
Uterine leiomyomas (ULs) are benign tumors occurring in the majority of reproductive aged women. Despite the high prevalence of these tumors, little is known about their etiology. A hallmark of ULs is the excessive deposition of extracellular matrix (ECM), primarily collagens. Collagens are known to modulate cell behavior and function singularly or through interactions with integrins and growth factor-mediated mitogenic pathways. To better understand the pathogenesis of ULs and the role of ECM collagens in their growth, we investigated the interaction of leiomyoma smooth muscle cells (LSMCs) with two different forms of collagen, non-polymerized collagen (monomeric) and polymerized collagen (fibrillar), in the absence or presence of platelet-derived growth factor (PDGF), an abundant growth factor in ULs. Primary cultures of human LSMCS from symptomatic patients were grown on these two different collagen matrices and their morphology, cytoskeletal organization, cellular proliferation, and signaling pathways were evaluated. Our results showed that LSMCs had distinct morphologies on the different collagen matrices and their basal as well as PDGF-stimulated proliferation varied on these matrices. These differences in proliferation were accompanied by changes in cell cycle progression and p21, an inhibitory cell cycle protein. In addition we found alterations in the phosphorylation of focal adhesion kinase, cytoskeletal reorganization, and activation of the mitogen activated protein kinase (MAPK) signaling pathway. In conclusion, our results demonstrate a direct effect of ECM on the proliferation of LSMCs through interplay between the collagen matrix and the PDGF-stimulated MAPK pathway. In addition, these findings will pave the way for identifying novel therapeutic approaches for ULs that target ECM proteins and their signaling pathways in ULs.
Collapse
Affiliation(s)
- Faezeh Koohestani
- Department of Animal Sciences, University of Illinois, Urbana, Illinois, United States of America
| | | | | | | | | | | |
Collapse
|
35
|
Abbonante V, Gruppi C, Rubel D, Gross O, Moratti R, Balduini A. Discoidin domain receptor 1 protein is a novel modulator of megakaryocyte-collagen interactions. J Biol Chem 2013; 288:16738-16746. [PMID: 23530036 DOI: 10.1074/jbc.m112.431528] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Growing evidence demonstrates that extracellular matrices regulate many aspects of megakaryocyte (MK) development; however, among the different extracellular matrix receptors, integrin α2β1 and glycoprotein VI are the only collagen receptors studied in platelets and MKs. In this study, we demonstrate the expression of the novel collagen receptor discoidin domain receptor 1 (DDR1) by human MKs at both mRNA and protein levels and provide evidence of DDR1 involvement in the regulation of MK motility on type I collagen through a mechanism based on the activity of SHP1 phosphatase and spleen tyrosine kinase (Syk). Specifically, we demonstrated that inhibition of DDR1 binding to type I collagen, preserving the engagement of the other collagen receptors, glycoprotein VI, α2β1, and LAIR-1, determines a decrease in MK migration due to the reduction in SHP1 phosphatase activity and consequent increase in the phosphorylation level of its main substrate Syk. Consistently, inhibition of Syk activity restored MK migration on type I collagen. In conclusion, we report the expression and function of a novel collagen receptor on human MKs, and we point out that an increasing level of complexity is necessary to better understand MK-collagen interactions in the bone marrow environment.
Collapse
Affiliation(s)
- Vittorio Abbonante
- Biotechnology Research Laboratories, Department of Molecular Medicine, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) San Matteo Foundation, University of Pavia, 27100 Pavia, Italy
| | - Cristian Gruppi
- Biotechnology Research Laboratories, Department of Molecular Medicine, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) San Matteo Foundation, University of Pavia, 27100 Pavia, Italy
| | - Diana Rubel
- Department of Nephrology and Rheumatology, University Medical Center Goettingen, 37075 Goettingen, Germany
| | - Oliver Gross
- Department of Nephrology and Rheumatology, University Medical Center Goettingen, 37075 Goettingen, Germany
| | - Remigio Moratti
- Biotechnology Research Laboratories, Department of Molecular Medicine, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) San Matteo Foundation, University of Pavia, 27100 Pavia, Italy
| | - Alessandra Balduini
- Biotechnology Research Laboratories, Department of Molecular Medicine, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) San Matteo Foundation, University of Pavia, 27100 Pavia, Italy; Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155.
| |
Collapse
|
36
|
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: 163] [Impact Index Per Article: 13.6] [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
|
37
|
Carafoli F, Hohenester E. Collagen recognition and transmembrane signalling by discoidin domain receptors. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2012; 1834:2187-94. [PMID: 23128141 PMCID: PMC4332414 DOI: 10.1016/j.bbapap.2012.10.014] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 10/23/2012] [Accepted: 10/25/2012] [Indexed: 12/20/2022]
Abstract
The discoidin domain receptors, DDR1 and DDR2, are two closely related receptor tyrosine kinases that are activated by triple-helical collagen in a slow and sustained manner. The DDRs have important roles in embryo development and their dysregulation is associated with human diseases, such as fibrosis, arthritis and cancer. The extracellular region of DDRs consists of a collagen-binding discoidin (DS) domain and a DS-like domain. The transmembrane region mediates the ligand-independent dimerisation of DDRs and is connected to the tyrosine kinase domain by an unusually long juxtamembrane domain. The major DDR binding site in fibrillar collagens is a GVMGFO motif (O is hydroxyproline), which is recognised by an amphiphilic trench at the top of the DS domain. How collagen binding leads to DDR activation is not understood. GVMGFO-containing triple-helical peptides activate DDRs with the characteristic slow kinetics, suggesting that the supramolecular structure of collagen is not required. Activation can be blocked allosterically by monoclonal antibodies that bind to the DS-like domain. Thus, collagen most likely causes a conformational change within the DDR dimer, which may lead to the formation of larger DDR clusters. This article is part of a Special Issue entitled: Emerging recognition and activation mechanisms of receptor tyrosine kinases.
Collapse
|
38
|
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: 286] [Impact Index Per Article: 22.0] [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
|
39
|
Lemeer S, Bluwstein A, Wu Z, Leberfinger J, Müller K, Kramer K, Kuster B. Phosphotyrosine mediated protein interactions of the discoidin domain receptor 1. J Proteomics 2011; 75:3465-77. [PMID: 22057045 DOI: 10.1016/j.jprot.2011.10.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 09/30/2011] [Accepted: 10/06/2011] [Indexed: 11/24/2022]
Abstract
The receptor tyrosine kinase DDR1 has been implicated in multiple human cancers and fibrosis and is targeted by the leukemia drug Gleevec. This suggests that DDR1 might be a new therapeutic target. However, further insight into the DDR1 signaling pathway is required in order to support its further development. Here, we investigated DDR1 proximal signaling by the analysis of protein-protein interactions using proteomic approaches. All known interactors of DDR1 were identified and localized to specific phosphotyrosine residues on the receptor. In addition, we identified numerous signaling proteins as new putative phosphotyrosine mediated interactors including RasGAP, SHIP1, SHIP2, STATs, PI3K and the SRC family kinases. Most of the new proteins contain SH2 and PTB domains and for all interactors we could directly point the site of interaction to specific phosphotyrosine residues on the receptor. The identified proteins have roles in the early steps of the signaling cascade, propagating the signal from the DDR1 receptor into the cell. The map of phosphotyrosine mediated interactors of DDR1 created in this study will serve as a starting point for functional investigations which will enhance our knowledge on the role of the DDR1 receptor in health and disease. This article is part of a Special Section entitled: Understanding genome regulation and genetic diversity by mass spectrometry.
Collapse
Affiliation(s)
- Simone Lemeer
- Chair of Proteomics and Bioanalytics, Technische Universität München, Emil Erlenmeyer Forum 5, 85354 Freising, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Chetoui N, El azreq MA, Boisvert M, Bergeron MÈ, Aoudjit F. Discoidin domain receptor 1 expression in activated T cells is regulated by the ERK MAP kinase signaling pathway. J Cell Biochem 2011; 112:3666-74. [DOI: 10.1002/jcb.23300] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
41
|
Deletion of discoidin domain receptor 2 does not affect smooth muscle cell adhesion, migration, or proliferation in response to type I collagen. Cardiovasc Pathol 2011; 21:214-8. [PMID: 21865059 DOI: 10.1016/j.carpath.2011.07.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 07/07/2011] [Accepted: 07/10/2011] [Indexed: 11/21/2022] Open
Abstract
Collagen receptors expressed on vascular smooth muscle cells include the discoidin domain receptors (DDR1 and DDR2). DDR1 is known to play important roles in mediating smooth muscle cell responses to vascular injury, including neointimal hyperplasia, but much less is known about the function of DDR2. In this study, we harvested smooth muscle cells from DDR2 wild-type and knockout mice and studied the cells using in vitro models of migration and growth. There were no significant differences in the ability of Ddr2(+/+) or Ddr2(-/-) smooth muscle cells to attach to, migrate, or proliferate on type I collagen. Furthermore, neither matrix metalloproteinase (MMP) 2 nor MMP-9 activity nor type I collagen expression was different between the cell types. We conclude that in vitro, endogenous DDR2 is not required for smooth muscle cell hyperplastic responses to collagen.
Collapse
|
42
|
Guerrot D, Kerroch M, Placier S, Vandermeersch S, Trivin C, Mael-Ainin M, Chatziantoniou C, Dussaule JC. Discoidin domain receptor 1 is a major mediator of inflammation and fibrosis in obstructive nephropathy. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:83-91. [PMID: 21640971 DOI: 10.1016/j.ajpath.2011.03.023] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 02/08/2011] [Accepted: 03/17/2011] [Indexed: 11/29/2022]
Abstract
The interactions between tubulointerstitial infiltrating cells and the extracellular matrix play an important role in regulating renal fibrosis. Discoidin domain receptor 1 (DDR1) is a nonintegrin tyrosine kinase receptor for collagen implicated in cell adhesion, proliferation, and extracellular matrix remodeling. We have previously demonstrated that transgenic mice lacking DDR1 are protected from hypertension-associated renal fibrosis. The purpose of this study was to determine the role of DDR1 in renal inflammation and fibrosis related to primitive tubulointerstitial injury. After 12 days of unilateral ureteral obstruction (UUO), kidney histopathologic and real-time quantitative PCR analyses were performed in DDR1(-/-) and wild-type mice. DDR1 expression was strongly increased in the obstructed kidney. Wild-type mice developed important perivascular and interstitial inflammation and fibrosis. In comparison, DDR1(-/-) mice displayed reduced accumulation of fibrillar collagen and transforming growth factor β expression. F4/80(+) cell count and proinflammatory cytokines were remarkably blunted in DDR1(-/-) obstructed kidneys. Leukocyte rolling and adhesion evaluated by intravital microscopy were not different between DDR1(-/-) and wild-type mice. Importantly, macrophages isolated from DDR1(-/-) mice presented similar M1/M2 polarization but displayed impaired migration in response to monocyte chemoattractant protein-1. Together, these data suggest that DDR1 plays an important role in the pathogenesis of renal disease via enhanced inflammation. Inhibition of DDR1 expression or activity may represent a novel therapeutic target against the progression of renal diseases.
Collapse
Affiliation(s)
- Dominique Guerrot
- INSERM UMR S 702, Hôpital Tenon, Paris, France; UPMC Paris 6 University, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
43
|
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
|
44
|
Castro-Sanchez L, Soto-Guzman A, Guaderrama-Diaz M, Cortes-Reynosa P, Salazar EP. Role of DDR1 in the gelatinases secretion induced by native type IV collagen in MDA-MB-231 breast cancer cells. Clin Exp Metastasis 2011; 28:463-77. [PMID: 21461859 DOI: 10.1007/s10585-011-9385-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 03/21/2011] [Indexed: 12/11/2022]
Abstract
Discoidin domain receptors (DDRs) are receptor tyrosine kinases that get activated by collagens in its native triple-helical form. In mammalian cells, DDR family consists of two members, namely DDR1 and DDR2, which mediates migration and proliferation of several cell types. DDR1 is activated by native type IV collagen and overexpressed in human breast cancer. Type IV collagen is the main component of basement membrane (BM), and the ability to degrade and penetrate BM is related with an increased potential for invasion and metastasis. Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases that collectively are capable of degrading all components of the extracellular matrix, including the BM. In breast cancer cells, denatured type IV collagen induces MMP-9 secretion and invasion. However, the role of DDR1 in the regulation of gelatinases (MMP-2 and -9) secretion and invasion in breast cancer cells remains to be studied. We demonstrate here that native type IV collagen induces MMP-2 and -9 secretions and invasion through a DDR1 and Src-dependent pathway, together with an increase of MMP-2 and -9-cell surface levels. MMP-2 and -9 secretions require PKC kinase activity, epidermal growth factor receptor (EGFR) activation, arachidonic acid (AA) production and AA metabolites in MDA-MB-231 breast cancer cells. In summary, our data demonstrate, for the first time, that DDR1 mediates MMP-2 and -9 secretions and invasion induced by native type IV collagen in MDA-MB-231 breast cancer cells.
Collapse
Affiliation(s)
- Luis Castro-Sanchez
- Departamento de Biologia Celular, Cinvestav-IPN, San Pedro Zacatenco, 07360, Mexico, DF, Mexico
| | | | | | | | | |
Collapse
|
45
|
Impaired dermal wound healing in discoidin domain receptor 2-deficient mice associated with defective extracellular matrix remodeling. FIBROGENESIS & TISSUE REPAIR 2011; 4:5. [PMID: 21288331 PMCID: PMC3044108 DOI: 10.1186/1755-1536-4-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Accepted: 02/02/2011] [Indexed: 11/27/2022]
Abstract
Background The wounding response relies on tightly regulated crosstalk between recruited fibroblasts and the collagenous extracellular matrix (ECM). Discoidin domain receptor 2 (DDR2) is a tyrosine kinase receptor for fibrillar collagen expressed during pathologic scarring, for example wound healing, arthritis and cancer. We have previously shown that DDR2 phosphorylation drives key wounding responses in skin fibroblasts including proliferation, chemotactic migration and secretion of both metalloproteinases and fibrillar collagen. In this study we compared healing of cutaneous wounds in DDR2+/+ and DDR2-/- mice and analyzed specific fibroblast responses. Results Cutaneous wound healing was significantly delayed in DDR2-/- mice compared with DDR2+/+ animals. Reduced α-smooth muscle actin (αSMA) expression and matrix metalloproteinase 2 (MMP2) activity in the DDR2-/- wound extracts indicated defective recruitment of skin fibroblasts. DDR2-/- wounds showed decreased tensile strength during healing, which correlated with a significant reduction in collagen content and defective collagen crosslinking. Non-wounded skin in DDR2-/- mice expressed less mRNA of the crosslinking enzymes lysyl oxidase (LOX), lysyl hydroxylase1 (LH1) and matricellular 'secreted protein, acidic and rich in cysteine' (SPARC; also known as osteonectin). Skin fibroblasts isolated from DDR2-/- mice displayed altered mRNA expression of a cluster of collagens, proteoglycans, integrins and MMPs that have been previously correlated with DDR2 expression, and reduced LOX, LH1 and SPARC mRNA levels and proteins. Stable reconstitution of wild-type DDR2 by retroviral infection restored LOX, LH1 and SPARC mRNA and protein levels in DDR2-/- fibroblasts. Contraction of collagen gels was reduced in DDR2-/- fibroblasts, accompanied by significantly reduced phosphorylated SrcY418. Inhibition of either LOX activity by β-aminoproprionitrile or MMP activity by N-[(2R)-2-(hydroxamido carbonylmethyl)-4-methylpentanoyl]-l-tryptophan methylamide (GM6001) reduced collagen gel contraction by skin fibroblasts after DDR2 induction with soluble collagen type I. Conclusions DDR2 contributes to skin fibroblast responses during tissue injury. Defective synthesis of collagen type I, crosslinking molecules and MMP2 predispose DDR2-/- mice to defective dermal wounding.
Collapse
|