1
|
Han X, Zhang C, Lei Q, Xu J, Zhou Y. Stiffness regulates extracellular matrix synthesis in fibroblasts by DDR1-TGF-β/STAT3 mechanotransduction axis. BIOMATERIALS ADVANCES 2025; 172:214240. [PMID: 40023083 DOI: 10.1016/j.bioadv.2025.214240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 02/10/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
For a long time, research on atherosclerosis (AS) has mainly focused on endothelial cells (ECs) and smooth muscle cells (SMCs) in blood vessels. Fibroblasts, however, being the major component in adventitia, little is known about their role. Fibroblasts are highly plastic cells, capable of undergoing phenotypic changes in response to various extracellular signals. Once activated, fibroblasts can promote fibrosis by altering the secretion of extracellular matrix (ECM). In this study, the effect of ECM stiffness on fibroblasts was investigated. Polyacrylamide (PA) gels with varying elastic moduli (1 kPa, 20 kPa and 100 kPa) were used as models for matrix stiffness. Human fibroblasts were cultured on these substrates, and their phenotypic and functional changes were examined. The data revealed that a collagen-binding receptor, Discoidin Domain Receptor 1 (DDR1), plays a central role in sensing mechanical stimuli from ECM. Matrix stiffness-induced phosphorylation of DDR1 suppresses the synthesis of ECM proteins in fibroblasts. The expression of ECM proteins on the 1 kPa substrate was significantly higher than that on the 20 kPa and 100 kPa substrates, while the phosphorylation level of DDR1 was notably reduced. After knocking out DDR1, the difference in ECM proteins expression among the three substrates with different stiffness levels disappeared. The signal transduction from DDR1 to ECM synthesis is mediated by the TGF-β/STAT3 signaling axis. Our study reveals how matrix stiffness regulates the synthesis of ECM in fibroblasts and paves the way for understanding the regulation of fibrotic process in the pathogenesis of AS.
Collapse
Affiliation(s)
- Xiaomei Han
- Department of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, PR China
| | - Chao Zhang
- Department of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, PR China
| | - Qian Lei
- Department of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, PR China
| | - Jin Xu
- Department of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, PR China
| | - Yue Zhou
- Department of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, PR China.
| |
Collapse
|
2
|
Ma Y, Gong H, Cheng L, Zhang D. Discoid Domain Receptors Signaling in Macrophages-Mediated Diseases. Int J Gen Med 2025; 18:907-926. [PMID: 39990299 PMCID: PMC11847422 DOI: 10.2147/ijgm.s487093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 01/18/2025] [Indexed: 02/25/2025] Open
Abstract
Macrophages, as a crucial component of the body's immune system, play a vital role in the onset, progression, and outcome of diseases. Discoidin domain receptors (DDRs), important members of the novel receptor tyrosine kinase superfamily, exhibit unique functions in macrophage physiology. Through interactions with the extracellular matrix, DDRs activate signaling pathways such as p38 MAPK and NF-κB, regulating macrophage adhesion, migration, and secretory functions, thereby influencing their behavior in diseases. Recent studies have indicated a direct correlation between DDRs and the progression of various diseases, including inflammation, cancer, and fibrosis. However, there remain numerous knowledge gaps regarding the specific mechanisms by which DDRs function in macrophage-mediated diseases. This article provides an in-depth summary of the regulatory mechanisms of DDRs on macrophages, detailing their modulatory roles in various diseases through macrophages and their underlying mechanisms. The aim is to offer new insights into biomedical therapies targeting DDRs and the development of novel drugs.
Collapse
Affiliation(s)
- Yaohui Ma
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Hang Gong
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Long Cheng
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Dekui Zhang
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| |
Collapse
|
3
|
Coelho NM, Riahi P, Wang Y, Ali A, Norouzi M, Kotlyar M, Jurisica I, McCulloch CA. The major vault protein integrates adhesion-driven signals to regulate collagen remodeling. Cell Signal 2024; 124:111447. [PMID: 39368789 DOI: 10.1016/j.cellsig.2024.111447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/21/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
DDR1 interacts with fibrillar collagen and can affect β1 integrin-dependent signaling, but the mechanism that mediates functional interactions between these two different receptors is not defined. We searched for molecules that link DDR1 and β1 integrin-dependent signaling in response to collagen binding. The activation of DDR1 by binding to fibrillar collagen reduced by 5-fold, β1 integrin-dependent ERK phosphorylation that leads to MMP1 expression. In contrast, pharmacological inhibition of DDR1 or culturing cells on fibronectin restored ERK phosphorylation and MMP1 expression mediated by the β1 integrin. A phospho-site screen indicated that collagen-induced DDR1 activation inhibited β1 integrin-dependent ERK signaling by regulating autophosphorylation of focal adhesion kinase (FAK). Immunoprecipitation, mass spectrometry, and protein-protein interaction mapping showed that while DDR1 and FAK do not interact directly, the major vault protein (MVP) binds DDR1 and FAK depending on the substrate. MVP associated with DDR1 in cells expressing β1 integrin that were cultured on collagen. Knockdown of MVP restored ERK activation and MMP1 expression in DDR1-expressing cells cultured on collagen. Immunostaining of invasive cancers in human colon showed colocalization of DDR1 with MVP. These data indicate that MVP interactions with DDR1 and FAK contribute to the regulation of β1 integrin-dependent signaling pathways that drive collagen degradation.
Collapse
Affiliation(s)
- Nuno M Coelho
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Pardis Riahi
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Yongqiang Wang
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Aiman Ali
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Masoud Norouzi
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Max Kotlyar
- Osteoarthritis Research Program, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, UHN, Toronto, ON, Canada
| | - Igor Jurisica
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada; Osteoarthritis Research Program, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, UHN, Toronto, ON, Canada; Departments of Medical Biophysics and Computer Science, University of Toronto, ON, Canada
| | | |
Collapse
|
4
|
Mierke CT. Mechanosensory entities and functionality of endothelial cells. Front Cell Dev Biol 2024; 12:1446452. [PMID: 39507419 PMCID: PMC11538060 DOI: 10.3389/fcell.2024.1446452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The endothelial cells of the blood circulation are exposed to hemodynamic forces, such as cyclic strain, hydrostatic forces, and shear stress caused by the blood fluid's frictional force. Endothelial cells perceive mechanical forces via mechanosensors and thus elicit physiological reactions such as alterations in vessel width. The mechanosensors considered comprise ion channels, structures linked to the plasma membrane, cytoskeletal spectrin scaffold, mechanoreceptors, and junctional proteins. This review focuses on endothelial mechanosensors and how they alter the vascular functions of endothelial cells. The current state of knowledge on the dysregulation of endothelial mechanosensitivity in disease is briefly presented. The interplay in mechanical perception between endothelial cells and vascular smooth muscle cells is briefly outlined. Finally, future research avenues are highlighted, which are necessary to overcome existing limitations.
Collapse
|
5
|
Wei Z, Li J, Zhong L, Yang D, Li W, Chen W, Zhou H, He Y, Song W, Wang B, Zeng L. DDR1 Drives Malignant Progression of Gastric Cancer by Suppressing HIF-1α Ubiquitination and Degradation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308395. [PMID: 39024501 PMCID: PMC11425230 DOI: 10.1002/advs.202308395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 06/07/2024] [Indexed: 07/20/2024]
Abstract
The extracellular matrix (ECM) has been demonstrated to be dysregulated and crucial for malignant progression in gastric cancer (GC), but the mechanism is not well understood. Here, that discoidin domain receptor 1 (DDR1), a principal ECM receptor, is recognized as a key driver of GC progression is reported. Mechanistically, DDR1 directly interacts with the PAS domain of hypoxia-inducible factor-1α (HIF-1α), suppresses its ubiquitination and subsequently strengthens its transcriptional regulation of angiogenesis. Additionally, DDR1 upregulation in GC cells promotes actin cytoskeleton reorganization by activating HIF-1α/ Ras Homolog Family Member A (RhoA)/Rho-associated protein kinase 1 (ROCK1) signaling, which in turn enhances the metastatic capacity. Pharmacological inhibition of DDR1 suppresses GC progression and angiogenesis in patient-derived xenograft (PDX) and organoid models. Taken together, this work first indicates the effects of the DDR1-HIF-1α axis on GC progression and reveals the related mechanisms, providing experimental evidence for DDR1 as a therapeutic target for GC.
Collapse
Affiliation(s)
- Zhewei Wei
- Department of Gastrointestinal SurgeryThe First Affiliated Hospital of Sun Yat‐sen University58 Zhongshan 2nd RoadGuangzhouGuangdong510080China
| | - Jin Li
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer ResearchScientific Research CenterThe BiobankThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityNo. 628 Zhenyuan RoadShenzhenGuangdong518107China
| | - Li Zhong
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer ResearchScientific Research CenterThe BiobankThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityNo. 628 Zhenyuan RoadShenzhenGuangdong518107China
| | - Dongjie Yang
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer ResearchScientific Research CenterThe BiobankThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityNo. 628 Zhenyuan RoadShenzhenGuangdong518107China
| | - Wuguo Li
- Laboratory Animal CenterThe First Affiliated HospitalSun Yat‐sen University58 Zhongshan 2nd RoadGuangzhou510080China
| | - Wei Chen
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer ResearchScientific Research CenterThe BiobankThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityNo. 628 Zhenyuan RoadShenzhenGuangdong518107China
| | - Hao Zhou
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer ResearchScientific Research CenterThe BiobankThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityNo. 628 Zhenyuan RoadShenzhenGuangdong518107China
| | - Yulong He
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer ResearchScientific Research CenterThe BiobankThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityNo. 628 Zhenyuan RoadShenzhenGuangdong518107China
| | - Wu Song
- Department of Gastrointestinal SurgeryThe First Affiliated Hospital of Sun Yat‐sen University58 Zhongshan 2nd RoadGuangzhouGuangdong510080China
| | - Boyan Wang
- Reproductive Medicine CenterThe First Affiliated Hospital of Sun Yat‐sen University58 Zhongshan 2nd RoadGuangzhouGuangdong510080China
| | - Leli Zeng
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer ResearchScientific Research CenterThe BiobankThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityNo. 628 Zhenyuan RoadShenzhenGuangdong518107China
| |
Collapse
|
6
|
Younesi FS, Miller AE, Barker TH, Rossi FMV, Hinz B. Fibroblast and myofibroblast activation in normal tissue repair and fibrosis. Nat Rev Mol Cell Biol 2024; 25:617-638. [PMID: 38589640 DOI: 10.1038/s41580-024-00716-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
The term 'fibroblast' often serves as a catch-all for a diverse array of mesenchymal cells, including perivascular cells, stromal progenitor cells and bona fide fibroblasts. Although phenotypically similar, these subpopulations are functionally distinct, maintaining tissue integrity and serving as local progenitor reservoirs. In response to tissue injury, these cells undergo a dynamic fibroblast-myofibroblast transition, marked by extracellular matrix secretion and contraction of actomyosin-based stress fibres. Importantly, whereas transient activation into myofibroblasts aids in tissue repair, persistent activation triggers pathological fibrosis. In this Review, we discuss the roles of mechanical cues, such as tissue stiffness and strain, alongside cell signalling pathways and extracellular matrix ligands in modulating myofibroblast activation and survival. We also highlight the role of epigenetic modifications and myofibroblast memory in physiological and pathological processes. Finally, we discuss potential strategies for therapeutically interfering with these factors and the associated signal transduction pathways to improve the outcome of dysregulated healing.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Andrew E Miller
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, USA
| | - Fabio M V Rossi
- School of Biomedical Engineering and Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Boris Hinz
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada.
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
7
|
Ho Thanh MT, Poudel A, Ameen S, Carroll B, Wu M, Soman P, Zhang T, Schwarz JM, Patteson AE. Vimentin promotes collective cell migration through collagen networks via increased matrix remodeling and spheroid fluidity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599259. [PMID: 38948855 PMCID: PMC11212918 DOI: 10.1101/2024.06.17.599259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The intermediate filament (IF) protein vimentin is associated with many diseases with phenotypes of enhanced cellular migration and aggressive invasion through the extracellular matrix (ECM) of tissues, but vimentin's role in in-vivo cell migration is still largely unclear. Vimentin is important for proper cellular adhesion and force generation, which are critical to cell migration; yet the vimentin cytoskeleton also hinders the ability of cells to squeeze through small pores in ECM, resisting migration. To identify the role of vimentin in collective cell migration, we generate spheroids of wide-type and vimentin-null mouse embryonic fibroblasts (mEFs) and embed them in a 3D collagen matrix. We find that loss of vimentin significantly impairs the ability of the spheroid to collectively expand through collagen networks and remodel the collagen network. Traction force analysis reveals that vimentin null spheroids exert less contractile force than their wild-type counterparts. In addition, spheroids made of mEFs with only vimentin unit length filaments (ULFs) exhibit similar behavior as vimentin-null spheroids, suggesting filamentous vimentin is required to promote 3D collective cell migration. We find the vimentin-mediated collective cell expansion is dependent on matrix metalloproteinase (MMP) degradation of the collagen matrix. Further, 3D vertex model simulation of spheroid and embedded ECM indicates that wild-type spheroids behave more fluid-like, enabling more active pulling and reconstructing the surrounding collagen network. Altogether, these results signify that VIF plays a critical role in enhancing migratory persistence in 3D matrix environments through MMP transportation and tissue fluidity.
Collapse
Affiliation(s)
- Minh Tri Ho Thanh
- Physics Department, Syracuse University; Syracuse, New York, USA
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
| | - Arun Poudel
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
- Biomedical and Chemical Engineering Department, Syracuse University; Syracuse, New York, USA
| | - Shabeeb Ameen
- Physics Department, Syracuse University; Syracuse, New York, USA
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
| | - Bobby Carroll
- Physics Department, Syracuse University; Syracuse, New York, USA
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
| | - M Wu
- Department of Biological and Environmental Engineering, Cornell University; Ithaca, New York, USA
| | - Pranav Soman
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
- Biomedical and Chemical Engineering Department, Syracuse University; Syracuse, New York, USA
| | - Tao Zhang
- Department of Polymer Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - J M Schwarz
- Physics Department, Syracuse University; Syracuse, New York, USA
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
- Indian Creek Farm, Ithaca, New York, USA
| | - Alison E Patteson
- Physics Department, Syracuse University; Syracuse, New York, USA
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
| |
Collapse
|
8
|
Wu D, Ding Z, Lu T, Chen Y, Zhang F, Lu S. DDR1-targeted therapies: current limitations and future potential. Drug Discov Today 2024; 29:103975. [PMID: 38580164 DOI: 10.1016/j.drudis.2024.103975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024]
Abstract
Discoidin domain receptor (DDR)-1 has a crucial role in regulating vital processes, including cell differentiation, proliferation, adhesion, migration, invasion, and matrix remodeling. Overexpression or activation of DDR1 in various pathological scenarios makes it a potential therapeutic target for the treatment of cancer, fibrosis, atherosclerosis, and neuropsychiatric, psychiatric, and neurodegenerative disorders. In this review, we summarize current therapeutic approaches targeting DDR1 from a medicinal chemistry perspective. Furthermore, we analyze factors other than issues of low selectivity and risk of resistance, contributing to the infrequent success of DDR1 inhibitors. The complex interplay between DDR1 and the extracellular matrix (ECM) necessitates additional validation, given that DDR1 might exhibit complex and synergistic interactions with other signaling molecules during ECM regulation. The mechanisms involved in DDR1 regulation in cancer and inflammation-related diseases also remain unknown.
Collapse
Affiliation(s)
- Donglin Wu
- School of Science, China Pharmaceutical University, Nanjing 211198, China
| | - Zihui Ding
- School of Science, China Pharmaceutical University, Nanjing 211198, China
| | - Tao Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery, China Pharmaceutical University, Nanjing 211198, China.
| | - Feng Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Shuai Lu
- School of Science, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
9
|
Liu M, Zhang J, Li X, Wang Y. Research progress of DDR1 inhibitors in the treatment of multiple human diseases. Eur J Med Chem 2024; 268:116291. [PMID: 38452728 DOI: 10.1016/j.ejmech.2024.116291] [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: 12/03/2023] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
Discoidin domain receptor 1 (DDR1) is a collagen-activated receptor tyrosine kinase (RTK) and plays pivotal roles in regulating cellular functions such as proliferation, differentiation, invasion, migration, and matrix remodeling. DDR1 is involved in the occurrence and progression of many human diseases, including cancer, fibrosis, and inflammation. Therefore, DDR1 represents a highly promising therapeutic target. Although no selective small-molecule inhibitors have reached clinical trials to date, many molecules have shown therapeutic effects in preclinical studies. For example, BK40143 has demonstrated significant promise in the therapy of neurodegenerative diseases. In this context, our perspective aims to provide an in-depth exploration of DDR1, encompassing its structure characteristics, biological functions, and disease relevance. Furthermore, we emphasize the importance of understanding the structure-activity relationship of DDR1 inhibitors and highlight the unique advantages of dual-target or multitarget inhibitors. We anticipate offering valuable insights into the development of more efficacious DDR1-targeted drugs.
Collapse
Affiliation(s)
- Mengying Liu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Neuro-system and Multimorbidity Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China
| | - Jifa Zhang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Neuro-system and Multimorbidity Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China
| | - Xiaoxue Li
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Neuro-system and Multimorbidity Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| |
Collapse
|
10
|
Zeltz C, Kusche-Gullberg M, Heljasvaara R, Gullberg D. Novel roles for cooperating collagen receptor families in fibrotic niches. Curr Opin Cell Biol 2023; 85:102273. [PMID: 37918273 DOI: 10.1016/j.ceb.2023.102273] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 11/04/2023]
Abstract
Recent data indicate that integrin and non-integrin collagen receptors cooperate in the fibrosis-specific microenvironment (i.e., the fibrotic niche). In certain tumor types, DDR1 can regulate the interaction with collagen III to regulate dormancy and metastasis, whereas in other tumor types, DDR1 can be shed and used to reorganize collagen. DDR1 expressed on tumor cells, together with DDR2 and α11β1 integrin expressed on cancer-associated fibroblasts, can increase tumor tissue stiffness. Integrin α1β1 and α2β1 are present on immune cells where they together with the immunosuppressive collagen receptor LAIR-1 can mediate binding to intratumor collagens. In summary, collagen-binding integrins together with DDRs, can create fibrillar collagen niches that act as traps to hinder immune cell trafficking into the tumor cell mass. Binding of collagens via LAIR-1 on immune cells in turn results in CD8+T-cell exhaustion. Continued studies of these complex interactions are needed for successful new stroma-based therapeutic interventions. In the current review, we will summarize recent data on collagen receptors with a special focus on their potential role in tumor fibrosis and highlight their collaborative roles in tumor fibrotic niches.
Collapse
Affiliation(s)
- Cédric Zeltz
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, 5009 Bergen, Norway
| | - Marion Kusche-Gullberg
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, 5009 Bergen, Norway
| | - Ritva Heljasvaara
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Donald Gullberg
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, 5009 Bergen, Norway.
| |
Collapse
|
11
|
Liu J, Zhao C, Xiao X, Li A, Liu Y, Zhao J, Fan L, Liang Z, Pang W, Yao W, Li W, Zhou J. Endothelial discoidin domain receptor 1 senses flow to modulate YAP activation. Nat Commun 2023; 14:6457. [PMID: 37833282 PMCID: PMC10576099 DOI: 10.1038/s41467-023-42341-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023] Open
Abstract
Mechanotransduction in endothelial cells is critical to maintain vascular homeostasis and can contribute to disease development, yet the molecules responsible for sensing flow remain largely unknown. Here, we demonstrate that the discoidin domain receptor 1 (DDR1) tyrosine kinase is a direct mechanosensor and is essential for connecting the force imposed by shear to the endothelial responses. We identify the flow-induced activation of endothelial DDR1 to be atherogenic. Shear force likely causes conformational changes of DDR1 ectodomain by unfolding its DS-like domain to expose the buried cysteine-287, whose exposure facilitates force-induced receptor oligomerization and phase separation. Upon shearing, DDR1 forms liquid-like biomolecular condensates and co-condenses with YWHAE, leading to nuclear translocation of YAP. Our findings establish a previously uncharacterized role of DDR1 in directly sensing flow, propose a conceptual framework for understanding upstream regulation of the YAP signaling, and offer a mechanism by which endothelial activation of DDR1 promotes atherosclerosis.
Collapse
Affiliation(s)
- Jiayu Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Chuanrong Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Xue Xiao
- National Laboratory of Biomacromolecules and Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Aohan Li
- National Laboratory for Condensed Matter Physics and Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yueqi Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Jianan Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Linwei Fan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Zhenhui Liang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Wei Pang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Weijuan Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Wei Li
- National Laboratory of Biomacromolecules and Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jing Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China.
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China.
| |
Collapse
|
12
|
Ezzo M, Hinz B. Novel approaches to target fibroblast mechanotransduction in fibroproliferative diseases. Pharmacol Ther 2023; 250:108528. [PMID: 37708995 DOI: 10.1016/j.pharmthera.2023.108528] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/09/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023]
Abstract
The ability of cells to sense and respond to changes in mechanical environment is vital in conditions of organ injury when the architecture of normal tissues is disturbed or lost. Among the various cellular players that respond to injury, fibroblasts take center stage in re-establishing tissue integrity by secreting and organizing extracellular matrix into stabilizing scar tissue. Activation, activity, survival, and death of scar-forming fibroblasts are tightly controlled by mechanical environment and proper mechanotransduction ensures that fibroblast activities cease after completion of the tissue repair process. Conversely, dysregulated mechanotransduction often results in fibroblast over-activation or persistence beyond the state of normal repair. The resulting pathological accumulation of extracellular matrix is called fibrosis, a condition that has been associated with over 40% of all deaths in the industrialized countries. Consequently, elements in fibroblast mechanotransduction are scrutinized for their suitability as anti-fibrotic therapeutic targets. We review the current knowledge on mechanically relevant factors in the fibroblast extracellular environment, cell-matrix and cell-cell adhesion structures, stretch-activated membrane channels, stress-regulated cytoskeletal structures, and co-transcription factors. We critically discuss the targetability of these elements in therapeutic approaches and their progress in pre-clinical and/or clinical trials to treat organ fibrosis.
Collapse
Affiliation(s)
- Maya Ezzo
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, and Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Boris Hinz
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, and Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
13
|
Ostrowska-Podhorodecka Z, Ali A, Norouzi M, Ding I, Abbasi S, Arora PD, Wong THF, Magalhaes M, McCulloch CA. Vimentin-mediated myosin 10 aggregation at tips of cell extensions drives MT1-MMP-dependent collagen degradation in colorectal cancer. FASEB J 2023; 37:e23097. [PMID: 37440280 DOI: 10.1096/fj.202300672r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/09/2023] [Accepted: 07/03/2023] [Indexed: 07/14/2023]
Abstract
Colorectal cancer (CRC) is a high prevalence adenocarcinoma with progressive increases in metastasis-related mortality, but the mechanisms governing the extracellular matrix (ECM) degradation important for metastasis in CRC are not well-defined. We investigated a functional relationship between vimentin (Vim) and myosin 10 (Myo10), and whether this relationship is associated with cancer progression. We tested the hypothesis that Vim regulates the aggregation of Myo10 at the tips of cell extensions, which increases membrane-type 1 matrix metalloproteinase (MT1-MMP)-associated local collagen proteolysis and ECM degradation. Analysis of CRC samples revealed colocalization of Vim with Myo10 and MT1-MMP in cell extensions adjacent to sites of collagen degradation, suggesting an association with local cell invasion. We analyzed cultured CRC cells and fibroblasts and found that Vim accelerates aggregation of Myo10 at cell tips, which increases the cell extension rate. Vim stabilizes the interaction of Myo10 with MT1-MMP, which in turn increases collagenolysis. Vim depletion reduced the aggregation of Myo10 at the cell extension tips and MT1-MMP-dependent collagenolysis. We propose that Vim interacts with Myo10, which in turn associates with MT1-MMP to facilitate the transport of these molecules to the termini of cell extensions and there enhance cancer invasion of soft connective tissues.
Collapse
Affiliation(s)
| | - Aiman Ali
- Oral Pathology and Oral Medicine, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Masoud Norouzi
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Isabel Ding
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Sevil Abbasi
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Pamma D Arora
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Timothy H F Wong
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Marco Magalhaes
- Oral Pathology and Oral Medicine, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
- Cancer Invasion and Metastasis Laboratory, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
- Dental and Maxillofacial Sciences Department, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | | |
Collapse
|
14
|
Zhu L, Liu L, Wang A, Liu J, Huang X, Zan T. Positive feedback loops between fibroblasts and the mechanical environment contribute to dermal fibrosis. Matrix Biol 2023; 121:1-21. [PMID: 37164179 DOI: 10.1016/j.matbio.2023.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/06/2023] [Accepted: 05/07/2023] [Indexed: 05/12/2023]
Abstract
Dermal fibrosis is characterized by excessive deposition of extracellular matrix in the dermis and affects millions of people worldwide and causes limited movement, disfigurement and psychological distress in patients. Fibroblast dysfunction of plays a central role in the pathogenesis of dermal fibrosis and is controlled by distinct factors. Recent studies support the hypothesis that fibroblasts can drive matrix deposition and stiffening, which in turn can exacerbate the functional dysregulation of fibroblasts. Ultimately, through a positive feedback loop, uncontrolled pathological fibrosis develops. This review aims to summarize the phenomenon and mechanism of the positive feedback loop in dermal fibrosis, and discuss potential therapeutic targets to help further elucidate the pathogenesis of dermal fibrosis and develop therapeutic strategies. In this review, fibroblast-derived compositional and structural changes in the ECM that lead to altered mechanical properties are briefly discussed. We focus on the mechanisms by which mechanical cues participate in dermal fibrosis progression. The mechanosensors discussed in the review include integrins, DDRs, proteoglycans, and mechanosensitive ion channels. The FAK, ERK, Akt, and Rho pathways, as well as transcription factors, including MRTF and YAP/TAZ, are also discussed. In addition, we describe stiffness-induced biological changes in the ECM on fibroblasts that contribute to the formation of a positive feedback loop. Finally, we discuss therapeutic strategies to treat the vicious cycle and present important suggestions for researchers conducting in-depth research.
Collapse
Affiliation(s)
- Liang Zhu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Lechen Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Aoli Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jinwen Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| | - Tao Zan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| |
Collapse
|
15
|
Liao X, Li X, Liu R. Extracellular-matrix mechanics regulate cellular metabolism: A ninja warrior behind mechano-chemo signaling crosstalk. Rev Endocr Metab Disord 2023; 24:207-220. [PMID: 36385696 DOI: 10.1007/s11154-022-09768-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/21/2022] [Indexed: 11/18/2022]
Abstract
Mechanical forces are the indispensable constituent of environmental cues, such as gravity, barometric pressure, vibration, and contact with bodies, which are involved in pattern and organogenesis, providing mechanical input to tissues and determining the ultimate fate of cells. Extracellular matrix (ECM) stiffness, the slow elastic force, carries the external physical force load onto the cell or outputs the internal force exerted by the cell and its neighbors into the environment. Accumulating evidence illustrates the pivotal role of ECM stiffness in the regulation of organogenesis, maintenance of tissue homeostasis, and the development of multiple diseases, which is largely fulfilled through its systematical impact on cellular metabolism. This review summarizes the establishment and regulation of ECM stiffness, the mechanisms underlying how ECM stiffness is sensed by cells and signals to modulate diverse cell metabolic pathways, and the physiological and pathological significance of the ECM stiffness-cell metabolism axis.
Collapse
Affiliation(s)
- Xiaoyu Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, 14, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China
| | - Xin Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, 14, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China
| | - Rui Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, 14, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
16
|
Gong H, Xu HM, Zhang DK. Focusing on discoidin domain receptors in premalignant and malignant liver diseases. Front Oncol 2023; 13:1123638. [PMID: 37007062 PMCID: PMC10050580 DOI: 10.3389/fonc.2023.1123638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/03/2023] [Indexed: 03/17/2023] Open
Abstract
Discoidin domain receptors (DDRs) are receptor tyrosine kinases on the membrane surface that bind to extracellular collagens, but they are rarely expressed in normal liver tissues. Recent studies have demonstrated that DDRs participate in and influence the processes underlying premalignant and malignant liver diseases. A brief overview of the potential roles of DDR1 and DDR2 in premalignant and malignant liver diseases is presented. DDR1 has proinflammatory and profibrotic benefits and promotes the invasion, migration and liver metastasis of tumour cells. However, DDR2 may play a pathogenic role in early-stage liver injury (prefibrotic stage) and a different role in chronic liver fibrosis and in metastatic liver cancer. These views are critically significant and first described in detail in this review. The main purpose of this review was to describe how DDRs act in premalignant and malignant liver diseases and their potential mechanisms through an in-depth summary of preclinical in vitro and in vivo studies. Our work aims to provide new ideas for cancer treatment and accelerate translation from bench to bedside.
Collapse
Affiliation(s)
| | | | - De-Kui Zhang
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
17
|
McGowan SE. Discoidin domain receptor-2 enhances secondary alveolar septation in mice by activating integrins and modifying focal adhesions. Am J Physiol Lung Cell Mol Physiol 2023; 324:L307-L324. [PMID: 36719983 PMCID: PMC9988528 DOI: 10.1152/ajplung.00169.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 12/20/2022] [Accepted: 01/09/2023] [Indexed: 02/02/2023] Open
Abstract
The extracellular matrix (ECM) of the pulmonary parenchyma must maintain the structural relationships among resident cells during the constant distortion imposed by respiration. This dictates that both the ECM and cells adapt to changes in shape, while retaining their attachment. Membrane-associated integrins and discoidin domain receptors (DDR) bind collagen and transmit signals to the cellular cytoskeleton. Although the contributions of DDR2 to collagen deposition and remodeling during osseous development are evident, it is unclear how DDR2 contributes to lung development. Using mice (smallie, Slie/Slie, DDR2Δ) bearing a spontaneous inactivating deletion within the DDR2 coding region, we observed a decrease in gas-exchange surface area and enlargement of alveolar ducts. Compared with fibroblasts isolated from littermate controls, DDR2Δ fibroblasts, spread more slowly, developed fewer lamellipodia, and were less responsive to the rigidity of neighboring collagen fibers. Activated β1-integrin (CD29) was reduced in focal adhesions (FA) of DDR2Δ fibroblasts, less phospho-zyxin localized to and fewer FA developed over ventral actin stress fibers, and the adhesions had a lower aspect ratio compared with controls. However, DDR2 deletion did not reduce cellular displacement of the ECM. Our findings indicate that DDR2, in concert with collagen-binding β1-integrins, regulates the timing and location of focal adhesion formation and how lung fibroblasts respond to ECM rigidity. Reduced rigidity sensing and mechano-responsiveness may contribute to the distortion of alveolar ducts, where the fiber cable-network is enriched and tensile forces are concentrated. Strategies targeting DDR2 could help guide fibroblasts to locations where tensile forces organize parenchymal repair.
Collapse
Affiliation(s)
- Stephen E McGowan
- Department of Veterans Affairs Research Service, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
| |
Collapse
|
18
|
Silva ME, Hernández-Andrade M, Abasolo N, Espinoza-Cruells C, Mansilla JB, Reyes CR, Aranda S, Esteban Y, Rodriguez-Calvo R, Martorell L, Muntané G, Rivera FJ, Vilella E. DDR1 and Its Ligand, Collagen IV, Are Involved in In Vitro Oligodendrocyte Maturation. Int J Mol Sci 2023; 24:ijms24021742. [PMID: 36675255 PMCID: PMC9866737 DOI: 10.3390/ijms24021742] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Discoidin domain receptor 1 (DDR1) is a tyrosine kinase receptor expressed in epithelial cells from different tissues in which collagen binding activates pleiotropic functions. In the brain, DDR1 is mainly expressed in oligodendrocytes (OLs), the function of which is unclear. Whether collagen can activate DDR1 in OLs has not been studied. Here, we assessed the expression of DDR1 during in vitro OL differentiation, including collagen IV incubation, and the capability of collagen IV to induce DDR1 phosphorylation. Experiments were performed using two in vitro models of OL differentiation: OLs derived from adult rat neural stem cells (NSCs) and the HOG16 human oligodendroglial cell line. Immunocytofluorescence, western blotting, and ELISA were performed to analyze these questions. The differentiation of OLs from NSCs was addressed using oligodendrocyte transcription factor 2 (Olig2) and myelin basic protein (MBP). In HOG16 OLs, collagen IV induced DDR1 phosphorylation through slow and sustained kinetics. In NSC-derived OLs, DDR1 was found in a high proportion of differentiating cells (MBP+/Olig2+), but its protein expression was decreased in later stages. The addition of collagen IV did not change the number of DDR1+/MBP+ cells but did accelerate OL branching. Here, we provide the first demonstration that collagen IV mediates the phosphorylation of DDR1 in HOG16 cells and that the in vitro co-expression of DDR1 and MBP is associated with accelerated branching during the differentiation of primary OLs.
Collapse
Affiliation(s)
- Maria Elena Silva
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile
- Institute of Pharmacy, Faculty of Sciences, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Matías Hernández-Andrade
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Nerea Abasolo
- Hospital Universitari Institut Pere Mata, Institut d’Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, 43206 Reus, Spain
| | - Cristóbal Espinoza-Cruells
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Josselyne B. Mansilla
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Carolina R. Reyes
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Selena Aranda
- Hospital Universitari Institut Pere Mata, Institut d’Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, 43206 Reus, Spain
- Centro de Investigación Biomédica en Red en Salud Mental, CIBERSAM-Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Yaiza Esteban
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, “Sant Joan” University Hospital, Institut d’Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), 28029 Madrid, Spain
| | - Ricardo Rodriguez-Calvo
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, “Sant Joan” University Hospital, Institut d’Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), 28029 Madrid, Spain
| | - Lourdes Martorell
- Hospital Universitari Institut Pere Mata, Institut d’Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, 43206 Reus, Spain
- Centro de Investigación Biomédica en Red en Salud Mental, CIBERSAM-Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Gerard Muntané
- Hospital Universitari Institut Pere Mata, Institut d’Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, 43206 Reus, Spain
- Centro de Investigación Biomédica en Red en Salud Mental, CIBERSAM-Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francisco J. Rivera
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile
- Translational Regenerative Neurobiology Group, Molecular and Integrative Biosciences Research Program (MIBS), Faculty of Biological and Environmental Sciences, University of Helsinki, 00014 Helsinki, Finland
- Correspondence: or (F.J.R.); (E.V.); Tel.: +358-50-598-8142 or +56-63-229-3011 (F.J.R.); +34-658-513-138 (E.V.)
| | - Elisabet Vilella
- Hospital Universitari Institut Pere Mata, Institut d’Investigació Sanitària Pere Virgili-CERCA, Universitat Rovira i Virgili, 43206 Reus, Spain
- Centro de Investigación Biomédica en Red en Salud Mental, CIBERSAM-Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: or (F.J.R.); (E.V.); Tel.: +358-50-598-8142 or +56-63-229-3011 (F.J.R.); +34-658-513-138 (E.V.)
| |
Collapse
|
19
|
Liu J, Wang J, Liu Y, Xie SA, Zhang J, Zhao C, Zhou Y, Pang W, Yao W, Peng Q, Wang X, Zhou J. Liquid-Liquid Phase Separation of DDR1 Counteracts the Hippo Pathway to Orchestrate Arterial Stiffening. Circ Res 2023; 132:87-105. [PMID: 36475898 DOI: 10.1161/circresaha.122.322113] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The Hippo-YAP (yes-associated protein) signaling pathway is modulated in response to various environmental cues. Activation of YAP in vascular smooth muscle cells conveys the extracellular matrix stiffness-induced changes in vascular smooth muscle cells phenotype and behavior. Recent studies have established a mechanoreceptive role of receptor tyrosine kinase DDR1 (discoidin domain receptor 1) in vascular smooth muscle cells. METHODS We conduced 5/6 nephrectomy in vascular smooth muscle cells-specific Ddr1-knockout mice, accompanied by pharmacological inhibition of the Hippo pathway kinase LATS1 (large tumor suppressor 1), to investigate DDR1 in YAP activation. We utilized polyacrylamide gels of varying stiffness or the DDR1 ligand, type I collagen, to stimulate the cells. We employed multiple molecular biological techniques to explore the role of DDR1 in controlling the Hippo pathway and to determine the mechanistic basis by which DDR1 exerts this effect. RESULTS We identified the requirement for DDR1 in stiffness/collagen-induced YAP activation. We uncovered that DDR1 underwent stiffness/collagen binding-stimulated liquid-liquid phase separation and co-condensed with LATS1 to inactivate LATS1. Mutagenesis experiments revealed that the transmembrane domain is responsible for DDR1 droplet formation. Purified DDR1 N-terminal and transmembrane domain was sufficient to drive its reversible condensation. Depletion of the DDR1 C-terminus led to failure in co-condensation with LATS1. Interaction between the DDR1 C-terminus and LATS1 competitively inhibited binding of MOB1 (Mps one binder 1) to LATS1 and thus the subsequent phosphorylation of LATS1. Introduction of the single-point mutants, histidine-745-proline and histidine-902-proline, to DDR1 on the C-terminus abolished the co-condensation. In mouse models, YAP activity was positively correlated with collagen I expression and arterial stiffness. LATS1 inhibition reactivated the YAP signaling in Ddr1-deficient vessels and abrogated the arterial softening effect of Ddr1 deficiency. CONCLUSIONS These findings identify DDR1 as a mediator of YAP activation by mechanical and chemical stimuli and demonstrate that DDR1 regulates LATS1 phosphorylation in an liquid-liquid phase separation-dependent manner.
Collapse
Affiliation(s)
- Jiayu Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., W.P., W.Y., J.Z.).,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., Y.Z., J.Z.).,National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., J.Z.)
| | - Jin Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., W.P., W.Y., J.Z.).,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., Y.Z., J.Z.).,National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., J.Z.).,Beijing Institute of Infectious Diseases, Beijing Key Laboratory of Emerging Infectious Diseases, National Center for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China (J.W.)
| | - Yueqi Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., W.P., W.Y., J.Z.).,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., Y.Z., J.Z.).,National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., J.Z.)
| | - Si-An Xie
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., W.P., W.Y., J.Z.).,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., Y.Z., J.Z.).,National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., J.Z.)
| | - Jianrui Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., W.P., W.Y., J.Z.).,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., Y.Z., J.Z.).,National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., J.Z.)
| | - Chuanrong Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., W.P., W.Y., J.Z.).,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., Y.Z., J.Z.).,National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., J.Z.)
| | - Yuan Zhou
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., Y.Z., J.Z.).,Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, China (Y.Z.)
| | - Wei Pang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., W.P., W.Y., J.Z.)
| | - Weijuan Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., W.P., W.Y., J.Z.)
| | - Qin Peng
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, China (Q.P.)
| | - Xiaohong Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, China (X.W.)
| | - Jing Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., W.P., W.Y., J.Z.).,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., Y.Z., J.Z.).,National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, China (J.L., J.W., Y.L., S.-A.X., J.Z., C.Z., J.Z.)
| |
Collapse
|
20
|
Schuster R, Younesi F, Ezzo M, Hinz B. The Role of Myofibroblasts in Physiological and Pathological Tissue Repair. Cold Spring Harb Perspect Biol 2023; 15:a041231. [PMID: 36123034 PMCID: PMC9808581 DOI: 10.1101/cshperspect.a041231] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Myofibroblasts are the construction workers of wound healing and repair damaged tissues by producing and organizing collagen/extracellular matrix (ECM) into scar tissue. Scar tissue effectively and quickly restores the mechanical integrity of lost tissue architecture but comes at the price of lost tissue functionality. Fibrotic diseases caused by excessive or persistent myofibroblast activity can lead to organ failure. This review defines myofibroblast terminology, phenotypic characteristics, and functions. We will focus on the central role of the cell, ECM, and tissue mechanics in regulating tissue repair by controlling myofibroblast action. Additionally, we will discuss how therapies based on mechanical intervention potentially ameliorate wound healing outcomes. Although myofibroblast physiology and pathology affect all organs, we will emphasize cutaneous wound healing and hypertrophic scarring as paradigms for normal tissue repair versus fibrosis. A central message of this review is that myofibroblasts can be activated from multiple cell sources, varying with local environment and type of injury, to either restore tissue integrity and organ function or create an inappropriate mechanical environment.
Collapse
Affiliation(s)
- Ronen Schuster
- Faculty of Dentistry, University of Toronto, Toronto, M5S 3E2 Ontario, Canada
| | - Fereshteh Younesi
- Faculty of Dentistry, University of Toronto, Toronto, M5S 3E2 Ontario, Canada
- Laboratory of Tissue Repair and Regeneration, Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| | - Maya Ezzo
- Faculty of Dentistry, University of Toronto, Toronto, M5S 3E2 Ontario, Canada
- Laboratory of Tissue Repair and Regeneration, Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, M5S 3E2 Ontario, Canada
- Laboratory of Tissue Repair and Regeneration, Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| |
Collapse
|
21
|
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
|
22
|
Borza CM, Bolas G, Pozzi A. Genetic and pharmacological tools to study the role of discoidin domain receptors in kidney disease. Front Pharmacol 2022; 13:1001122. [PMID: 36249782 PMCID: PMC9554349 DOI: 10.3389/fphar.2022.1001122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Following injury the kidney undergoes a repair process, which results in replacement of the injured tissue with little evidence of damage. However, repetitive injuries or inability of the kidney to stop the repair process result in abnormal deposition of extracellular matrix (ECM) components leading to fibrosis and organ dysfunction. The synthesis/degradation of ECM components is finely regulated by several factors, including discoidin domain receptors (DDRs). These are receptor tyrosine kinases that are activated by collagens. Upon activation, DDRs control several cell functions that, when exacerbated, contribute to kidney injury and fibrosis. DDRs are undetectable in healthy kidney, but become rapidly upregulated in several kidney fibrotic conditions, thus making them attractive anti-fibrotic targets. DDRs contribute to kidney injury and fibrosis by promoting apoptosis of injured kidney cells, stimulating the production of pro-inflammatory cytokines, and regulating the production of ECM components. They achieve these effects by activating canonical intracellular molecules or by directly interacting with nuclear chromatin and promoting the transcription of pro-fibrotic genes. The goal of this review is to highlight canonical and non-canonical mechanisms whereby DDRs contribute to kidney injury/fibrosis. This review will summarize key findings obtained using cells and mice lacking DDRs and it will discuss the discovery and development of targeted DDR small molecule- and antisense-based inhibitors. Understanding the molecular mechanisms whereby DDRs control kidney injury and fibrosis might enable us to not only develop more selective and potent inhibitors, but to also determine when DDR inhibition needs to be achieved to prevent and/or halt the development of kidney fibrosis.
Collapse
Affiliation(s)
- Corina M. Borza
- Department of Medicine (Division of Nephrology), Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Gema Bolas
- Department of Medicine (Division of Nephrology), Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Ambra Pozzi
- Department of Medicine (Division of Nephrology), Vanderbilt University School of Medicine, Nashville, TN, United States
- Veterans Affairs Hospitals, Nashville, TN, United States
| |
Collapse
|
23
|
Sirvent A, Espie K, Papadopoulou E, Naim D, Roche S. New functions of DDR1 collagen receptor in tumor dormancy, immune exclusion and therapeutic resistance. Front Oncol 2022; 12:956926. [PMID: 35936735 PMCID: PMC9355703 DOI: 10.3389/fonc.2022.956926] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/28/2022] [Indexed: 01/22/2023] Open
Abstract
The tumor microenvironment facilitates cancer progression and therapeutic resistance. Tumor collagens and their architecture play an essential role in this process. However, little is known about the mechanisms by which tumor cells sense and respond to this extracellular matrix environment. Recently, the Discoidin Domain Receptor 1 (DDR1), a collagen receptor and tyrosine kinase has emerged as an important player in this malignant process, although the underlying signaling mechanisms remain unclear. Here, we review new DDR1 functions in tumor dormancy following dissemination, immune exclusion and therapeutic resistance induced by stromal collagens deposition. We also discuss the signaling mechanisms behind these tumor activities and the therapeutic strategies aiming at targeting these collagens-dependent tumor responses.
Collapse
Affiliation(s)
| | | | | | | | - Serge Roche
- *Correspondence: Serge Roche, ; Audrey Sirvent,
| |
Collapse
|
24
|
Ngai D, Mohabeer AL, Mao A, Lino M, Bendeck MP. Stiffness-Responsive Feedback Autoregulation of DDR1 Expression is Mediated by a DDR1-YAP/TAZ Axis. Matrix Biol 2022; 110:129-140. [PMID: 35562016 DOI: 10.1016/j.matbio.2022.05.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 05/02/2022] [Accepted: 05/05/2022] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Increased matrix stiffness is sensed by the collagen-binding receptor tyrosine kinase discoidin domain receptor 1 (DDR1). We have previously shown that DDR1 stimulates a positive feedback loop to increase its own expression in vascular smooth muscle cells (VSMCs). The transcriptional co-factors YAP/TAZ are stiffness sensing molecules that have not previously been investigated in DDR1 signaling. Here, we test the hypothesis that DDR1 signals through YAP/TAZ to auto-regulate its own expression. APPROACH AND RESULTS We used vascular smooth muscle cells (VSMCs) from wild-type and DDR1 knockout mice stimulated with collagen and/or substrates of different stiffness. We show that DDR1 controls YAP/TAZ nuclear localization and activity, whereas knockdown of YAP/TAZ attenuates DDR1 expression. In response to increased substrate stiffness, collagen stimulation, or RhoA activation, YAP/TAZ translocate to the nucleus and bind to chromatin. Finally, collagen stimulation promotes increased YAP/TAZ association with the Ddr1 promoter. CONCLUSIONS These findings reveal the mechanism by which DDR1 regulates YAP/TAZ activity which can then mediate positive feedback regulation of DDR1 expression by promoting transcription of the DDR1 gene.
Collapse
Affiliation(s)
- David Ngai
- Department of Laboratory Medicine and Pathobiology, University of Toronto; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto
| | - Amanda L Mohabeer
- Department of Laboratory Medicine and Pathobiology, University of Toronto; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto
| | - Amanda Mao
- Department of Laboratory Medicine and Pathobiology, University of Toronto; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto
| | - Marsel Lino
- Department of Laboratory Medicine and Pathobiology, University of Toronto; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto
| | - Michelle P Bendeck
- Department of Laboratory Medicine and Pathobiology, University of Toronto; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto.
| |
Collapse
|
25
|
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
|
26
|
Borza CM, Bolas G, Zhang X, Browning Monroe MB, Zhang MZ, Meiler J, Skwark MJ, Harris RC, Lapierre LA, Goldenring JR, Hook M, Rivera J, Brown KL, Leitinger B, Tyska MJ, Moser M, Böttcher RT, Zent R, Pozzi A. The Collagen Receptor Discoidin Domain Receptor 1b Enhances Integrin β1-Mediated Cell Migration by Interacting With Talin and Promoting Rac1 Activation. Front Cell Dev Biol 2022; 10:836797. [PMID: 35309920 PMCID: PMC8928223 DOI: 10.3389/fcell.2022.836797] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/04/2022] [Indexed: 01/17/2023] Open
Abstract
Integrins and discoidin domain receptors (DDRs) 1 and 2 promote cell adhesion and migration on both fibrillar and non fibrillar collagens. Collagen I contains DDR and integrin selective binding motifs; however, the relative contribution of these two receptors in regulating cell migration is unclear. DDR1 has five isoforms (DDR1a-e), with most cells expressing the DDR1a and DDR1b isoforms. We show that human embryonic kidney 293 cells expressing DDR1b migrate more than DDR1a expressing cells on DDR selective substrata as well as on collagen I in vitro. In addition, DDR1b expressing cells show increased lung colonization after tail vein injection in nude mice. DDR1a and DDR1b differ from each other by an extra 37 amino acids in the DDR1b cytoplasmic domain. Interestingly, these 37 amino acids contain an NPxY motif which is a central control module within the cytoplasmic domain of β integrins and acts by binding scaffold proteins, including talin. Using purified recombinant DDR1 cytoplasmic tail proteins, we show that DDR1b directly binds talin with higher affinity than DDR1a. In cells, DDR1b, but not DDR1a, colocalizes with talin and integrin β1 to focal adhesions and enhances integrin β1-mediated cell migration. Moreover, we show that DDR1b promotes cell migration by enhancing Rac1 activation. Mechanistically DDR1b interacts with the GTPase-activating protein (GAP) Breakpoint cluster region protein (BCR) thus reducing its GAP activity and enhancing Rac activation. Our study identifies DDR1b as a major driver of cell migration and talin and BCR as key players in the interplay between integrins and DDR1b in regulating cell migration.
Collapse
Affiliation(s)
- Corina M. Borza
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN, United States
| | - Gema Bolas
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN, United States
| | - Xiuqi Zhang
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN, United States
| | | | - Ming-Zhi Zhang
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN, United States
| | - Jens Meiler
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
- Leipzig University Medical School, Institute for Drug Discovery, Leipzig, Germany
| | - Marcin J. Skwark
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - Raymond C. Harris
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN, United States
| | - Lynne A. Lapierre
- Department of Surgery, Vanderbilt University, Nashville, TN, United States
- Veterans Affairs Hospital, Nashville, TN, United States
| | - James R. Goldenring
- Department of Surgery, Vanderbilt University, Nashville, TN, United States
- Veterans Affairs Hospital, Nashville, TN, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
| | - Magnus Hook
- Texas A&M Health Science Center Institute of Biosciences and Technology, Houston, TX, United States
| | - Jose Rivera
- Texas A&M Health Science Center Institute of Biosciences and Technology, Houston, TX, United States
| | - Kyle L. Brown
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN, United States
| | - Birgit Leitinger
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Matthew J. Tyska
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
| | - Markus Moser
- Department for Molecular Medicine, Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | - Ralph T. Böttcher
- Department for Molecular Medicine, Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | - Roy Zent
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN, United States
- Veterans Affairs Hospital, Nashville, TN, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
| | - Ambra Pozzi
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN, United States
- Veterans Affairs Hospital, Nashville, TN, United States
| |
Collapse
|
27
|
Molladavoodi S, DeWitte‐Orr SJ, Gregory DE. An in vitro 3D annulus fibrosus cell culture model with type I collagen: An examination of cell-matrix interactions. JOR Spine 2022; 5:e1193. [PMID: 35386752 PMCID: PMC8966884 DOI: 10.1002/jsp2.1193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 11/07/2022] Open
Abstract
Background Disorders of the intervertebral disc (IVD) are widely known to result in low back pain; one of the most common debilitating conditions worldwide. As a multifaceted condition, both inflammatory environment and mechanical factors can play a crucial role in IVD damage, and in particular, in the annulus fibrosus (AF), the highly collagenous outer ring of the IVD. As a result, a better understanding of how cells from the IVD, and specifically the AF, interact and respond to their environment is imperative. Goal The goal of this study is to use collagen type I as an in vitro three-dimensional extracellular matrix for AF cells of IVD and briefly examine both the cellular and mechanical effect of exposure to an inflammatory stimulant. Methods We utilized type I collagen as a 3D in vitro model material for culturing AF cells of Sprague Dawley rat tail IVDs. Results We showed that the cultured cells are viable and metabolically active; these cells also induced a distinct and significant contraction on their collagen matrix. Furthermore, to demonstrate potential versatility of our model our model and its versatility, we used lipopolysaccharide (LPS), as a known inflammatory stimulant in IVDs, to manipulate the cells and their interaction. LPS treatment resulted in detectable changes to the contraction cells induced on the collagen matrix and affected the mechanical properties of these constructs.
Collapse
Affiliation(s)
- Sara Molladavoodi
- Department of Kinesiology and Physical EducationWilfrid Laurier UniversityWaterlooOntarioCanada
| | - Stephanie J. DeWitte‐Orr
- Department of Health SciencesWilfrid Laurier UniversityWaterlooOntarioCanada
- Department of BiologyWilfrid Laurier UniversityWaterlooOntarioCanada
| | - Diane E. Gregory
- Department of Kinesiology and Physical EducationWilfrid Laurier UniversityWaterlooOntarioCanada
- Department of Health SciencesWilfrid Laurier UniversityWaterlooOntarioCanada
| |
Collapse
|
28
|
Wang AY, Coelho NM, Arora PD, Wang Y, Eymael D, Ji C, Wang Q, Lee W, Xu J, Kapus A, Carneiro KMM, McCulloch CA. DDR1 associates with TRPV4 in cell-matrix adhesions to enable calcium-regulated myosin activity and collagen compaction. J Cell Physiol 2022; 237:2451-2468. [PMID: 35150133 DOI: 10.1002/jcp.30696] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 01/10/2022] [Accepted: 01/27/2022] [Indexed: 11/10/2022]
Abstract
Tissue fibrosis manifests as excessive deposition of compacted, highly aligned collagen fibrils, which interfere with organ structure and function. Cells in collagen-rich lesions often exhibit marked overexpression of discoidin domain receptor 1 (DDR1), which is linked to increased collagen compaction through the association of DDR1 with the Ca2+ -dependent nonmuscle myosin IIA (NMIIA). We examined the functional relationship between DDR1 and the transient receptor potential vanilloid type 4 (TRPV4) channel, a Ca2+ -permeable ion channel that is implicated in collagen compaction. Fibroblasts expressing high levels of DDR1 were used to model cells in lesions with collagen compaction. In these cells, the expression of the β1 integrin was deleted to simplify studies of DDR1 function. Compared with DDR1 wild-type cells, high DDR1 expression was associated with increased Ca2+ influx through TRPV4, enrichment of TRPV4 in collagen adhesions, and enhanced contractile activity mediated by NMIIA. At cell adhesion sites to collagen, DDR1 associated with TRPV4, which enhanced DDR1-mediated collagen alignment and compaction. We conclude that DDR1 regulates Ca2+ influx through the TRPV4 channel to promote critical, DDR1-mediated processes that are important in lesions with collagen compaction and alignment.
Collapse
Affiliation(s)
- Andrew Y Wang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Nuno M Coelho
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Pamma D Arora
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Yongqiang Wang
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Denise Eymael
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Chenfan Ji
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Qin Wang
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Wilson Lee
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Jessica Xu
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Andras Kapus
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital and Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Karina M M Carneiro
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Christopher A McCulloch
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
29
|
Borza CM, Bolas G, Bock F, Zhang X, Akabogu FC, Zhang MZ, de Caestecker M, Yang M, Yang H, Lee E, Gewin L, Fogo AB, McDonald WH, Zent R, Pozzi A. DDR1 contributes to kidney inflammation and fibrosis by promoting the phosphorylation of BCR and STAT3. JCI Insight 2022; 7:e150887. [PMID: 34941574 PMCID: PMC8855801 DOI: 10.1172/jci.insight.150887] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 12/21/2021] [Indexed: 11/25/2022] Open
Abstract
Discoidin domain receptor 1 (DDR1), a receptor tyrosine kinase activated by collagen, contributes to chronic kidney disease. However, its role in acute kidney injury and subsequent development of kidney fibrosis is not clear. Thus, we performed a model of severe ischemia/reperfusion-induced acute kidney injury that progressed to kidney fibrosis in WT and Ddr1-null mice. We showed that Ddr1-null mice had reduced acute tubular injury, inflammation, and tubulointerstitial fibrosis with overall decreased renal monocyte chemoattractant protein (MCP-1) levels and STAT3 activation. We identified breakpoint cluster region (BCR) protein as a phosphorylated target of DDR1 that controls MCP-1 production in renal proximal tubule epithelial cells. DDR1-induced BCR phosphorylation or BCR downregulation increased MCP-1 secretion, suggesting that BCR negatively regulates the levels of MCP-1. Mechanistically, phosphorylation or downregulation of BCR increased β-catenin activity and in turn MCP-1 production. Finally, we showed that DDR1-mediated STAT3 activation was required to stimulate the secretion of TGF-β. Thus, DDR1 contributes to acute and chronic kidney injury by regulating BCR and STAT3 phosphorylation and in turn the production of MCP-1 and TGF-β. These findings identify DDR1 an attractive therapeutic target for ameliorating both proinflammatory and profibrotic signaling in kidney disease.
Collapse
Affiliation(s)
- Corina M. Borza
- Department of Medicine, Division of Nephrology and Hypertension, and
| | - Gema Bolas
- Department of Medicine, Division of Nephrology and Hypertension, and
| | - Fabian Bock
- Department of Medicine, Division of Nephrology and Hypertension, and
| | - Xiuqi Zhang
- Department of Medicine, Division of Nephrology and Hypertension, and
| | - Favour C. Akabogu
- Department of Medicine, Division of Nephrology and Hypertension, and
| | - Ming-Zhi Zhang
- Department of Medicine, Division of Nephrology and Hypertension, and
| | | | - Min Yang
- Department of Medicine, Division of Nephrology and Hypertension, and
| | - Haichun Yang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Ethan Lee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Leslie Gewin
- Division of Nephrology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Agnes B. Fogo
- Department of Medicine, Division of Nephrology and Hypertension, and
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - W. Hayes McDonald
- Proteomics Laboratory, Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee, USA
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Roy Zent
- Department of Medicine, Division of Nephrology and Hypertension, and
- Department of Veterans Affairs, Nashville, Nashville, Tennessee, USA
| | - Ambra Pozzi
- Department of Medicine, Division of Nephrology and Hypertension, and
- Department of Veterans Affairs, Nashville, Nashville, Tennessee, USA
| |
Collapse
|
30
|
Sterczała B, Chwiłkowska A, Szwedowicz U, Kobielarz M, Chwiłkowski B, Dominiak M. Impact of APRF+ in Combination with Autogenous Fibroblasts on Release Growth Factors, Collagen, and Proliferation and Migration of Gingival Fibroblasts: An In Vitro Study. MATERIALS 2022; 15:ma15030796. [PMID: 35160741 PMCID: PMC8836484 DOI: 10.3390/ma15030796] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 01/01/2023]
Abstract
The present study aimed to compare the action of advanced platelet-rich fibrin (A-PRF+) alone with the action of A-PRF+ combined with autologous gingival fibroblasts. The components released from A-PRF+ conditioned with autogenous fibroblasts that were quantified in the study were fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), trans-forming growth factor-beta1 and 2 (TGFβ1 and TGFβ2), and soluble collagen. A-PRF+ combined with fibroblasts demonstrated significantly higher values of released VEGF at every time point and, after 7 days, significantly higher values of released TGFβ2. A viability test after 72 h showed a significant increase in proliferation fibroblasts after exposition to the factors released from A-PRF+ combined with fibroblasts. Similarly, the degree of wound closure after 48 h was significantly higher for the factors released from A-RRF+ alone and the factors released from A-RRF+ combined with fibroblasts. These results imply that platelet-rich fibrin (PRF) enhanced with fibroblasts can be an alternative method of connective tissue transplantation.
Collapse
Affiliation(s)
- Barbara Sterczała
- Dental Surgery Department, Wroclaw Medical University, 50-425 Wroclaw, Poland;
- Correspondence: (B.S.); (A.C.); Tel.: +48-502-932-269 (B.S.)
| | - Agnieszka Chwiłkowska
- Department of Molecular and Cell Biology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
- Correspondence: (B.S.); (A.C.); Tel.: +48-502-932-269 (B.S.)
| | - Urszula Szwedowicz
- Department of Molecular and Cell Biology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Magdalena Kobielarz
- Department of Mechanics, Materials and Biomedical Engineering, Wroclaw University of Science and Technology, 50-371 Wroclaw, Poland;
| | - Bartłomiej Chwiłkowski
- Department of Applied Mathematics, Faculty of Pure and Applied Mathematics, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland;
| | - Marzena Dominiak
- Dental Surgery Department, Wroclaw Medical University, 50-425 Wroclaw, Poland;
| |
Collapse
|
31
|
Di Martino JS, Nobre AR, Mondal C, Taha I, Farias EF, Fertig EJ, Naba A, Aguirre-Ghiso JA, Bravo-Cordero JJ. A tumor-derived type III collagen-rich ECM niche regulates tumor cell dormancy. NATURE CANCER 2022; 3:90-107. [PMID: 35121989 PMCID: PMC8818089 DOI: 10.1038/s43018-021-00291-9] [Citation(s) in RCA: 157] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/21/2021] [Indexed: 04/14/2023]
Abstract
Cancer cells disseminate and seed in distant organs, where they can remain dormant for many years before forming clinically detectable metastases. Here we studied how disseminated tumor cells sense and remodel the extracellular matrix (ECM) to sustain dormancy. ECM proteomics revealed that dormant cancer cells assemble a type III collagen-enriched ECM niche. Tumor-derived type III collagen is required to sustain tumor dormancy, as its disruption restores tumor cell proliferation through DDR1-mediated STAT1 signaling. Second-harmonic generation two-photon microscopy further revealed that the dormancy-to-reactivation transition is accompanied by changes in type III collagen architecture and abundance. Analysis of clinical samples revealed that type III collagen levels were increased in tumors from patients with lymph node-negative head and neck squamous cell carcinoma compared to patients who were positive for lymph node colonization. Our data support the idea that the manipulation of these mechanisms could serve as a barrier to metastasis through disseminated tumor cell dormancy induction.
Collapse
Affiliation(s)
- Julie S Di Martino
- Division of Hematology and Oncology, Department of Medicine, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ana Rita Nobre
- Division of Hematology and Oncology, Department of Medicine and Department of Otolaryngology, Department of Oncological Sciences, Black Family Stem Cell Institute, Precision Immunology Institute, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Abel Salazar Biomedical Sciences Institute, University of Porto, Porto, Portugal
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chandrani Mondal
- Division of Hematology and Oncology, Department of Medicine, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Isra Taha
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Eduardo F Farias
- Division of Hematology and Oncology, Department of Medicine, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elana J Fertig
- Departments of Oncology, Applied Mathematics and Statistics and Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Julio A Aguirre-Ghiso
- Division of Hematology and Oncology, Department of Medicine and Department of Otolaryngology, Department of Oncological Sciences, Black Family Stem Cell Institute, Precision Immunology Institute, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Cell Biology, Cancer Dormancy and Tumor Microenvironment Institute, Gruss Lipper Biophotonics Center, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jose Javier Bravo-Cordero
- Division of Hematology and Oncology, Department of Medicine, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
32
|
Berestjuk I, Lecacheur M, Carminati A, Diazzi S, Rovera C, Prod'homme V, Ohanna M, Popovic A, Mallavialle A, Larbret F, Pisano S, Audebert S, Passeron T, Gaggioli C, Girard CA, Deckert M, Tartare-Deckert S. Targeting Discoidin Domain Receptors DDR1 and DDR2 overcomes matrix-mediated tumor cell adaptation and tolerance to BRAF-targeted therapy in melanoma. EMBO Mol Med 2021; 14:e11814. [PMID: 34957688 PMCID: PMC8819497 DOI: 10.15252/emmm.201911814] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/04/2022] Open
Abstract
Resistance to BRAF/MEK inhibitor therapy in BRAFV600‐mutated advanced melanoma remains a major obstacle that limits patient benefit. Microenvironment components including the extracellular matrix (ECM) can support tumor cell adaptation and tolerance to targeted therapy; however, the underlying mechanisms remain poorly understood. Here, we investigated the process of matrix‐mediated drug resistance (MMDR) in response to BRAFV600 pathway inhibition in melanoma. We demonstrate that physical and structural cues from fibroblast‐derived ECM abrogate anti‐proliferative responses to BRAF/MEK inhibition. MMDR is mediated by drug‐induced linear clustering of phosphorylated DDR1 and DDR2, two tyrosine kinase collagen receptors. Depletion and pharmacological targeting of DDR1 and DDR2 overcome ECM‐mediated resistance to BRAF‐targeted therapy. In xenografts, targeting DDR with imatinib enhances BRAF inhibitor efficacy, counteracts drug‐induced collagen remodeling, and delays tumor relapse. Mechanistically, DDR‐dependent MMDR fosters a targetable pro‐survival NIK/IKKα/NF‐κB2 pathway. These findings reveal a novel role for a collagen‐rich matrix and DDR in tumor cell adaptation and resistance. They also provide important insights into environment‐mediated drug resistance and a preclinical rationale for targeting DDR signaling in combination with targeted therapy in melanoma.
Collapse
Affiliation(s)
- Ilona Berestjuk
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Team MicroCan, Nice, France
| | - Margaux Lecacheur
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Team MicroCan, Nice, France
| | - Alexandrine Carminati
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Team MicroCan, Nice, France
| | - Serena Diazzi
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Team MicroCan, Nice, France
| | - Christopher Rovera
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Team MicroCan, Nice, France
| | - Virginie Prod'homme
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Team MicroCan, Nice, France
| | - Mickael Ohanna
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Team MicroCan, Nice, France
| | - Ana Popovic
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Team MicroCan, Nice, France
| | - Aude Mallavialle
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Team MicroCan, Nice, France
| | - Frédéric Larbret
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Team MicroCan, Nice, France
| | - Sabrina Pisano
- Université Côte d'Azur, CNRS, INSERM, IRCAN, Nice, France
| | - Stéphane Audebert
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Thierry Passeron
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, Department of Dermatology, Nice, France
| | | | - Christophe A Girard
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Team MicroCan, Nice, France
| | - Marcel Deckert
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Team MicroCan, Nice, France
| | - Sophie Tartare-Deckert
- Université Côte d'Azur, INSERM, C3M, Nice, France.,Equipe labellisée Ligue Contre le Cancer, Team MicroCan, Nice, France
| |
Collapse
|
33
|
Muntz I, Fenu M, van Osch GJVM, Koenderink G. The role of cell-matrix interactions in connective tissue mechanics. Phys Biol 2021; 19. [PMID: 34902848 DOI: 10.1088/1478-3975/ac42b8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/13/2021] [Indexed: 11/12/2022]
Abstract
Living tissue is able to withstand large stresses in everyday life, yet it also actively adapts to dynamic loads. This remarkable mechanical behaviour emerges from the interplay between living cells and their non-living extracellular environment. Here we review recent insights into the biophysical mechanisms involved in the reciprocal interplay between cells and the extracellular matrix and how this interplay determines tissue mechanics, with a focus on connective tissues. We first describe the roles of the main macromolecular components of the extracellular matrix in regards to tissue mechanics. We then proceed to highlight the main routes via which cells sense and respond to their biochemical and mechanical extracellular environment. Next we introduce the three main routes via which cells can modify their extracellular environment: exertion of contractile forces, secretion and deposition of matrix components, and matrix degradation. Finally we discuss how recent insights in the mechanobiology of cell-matrix interactions are furthering our understanding of the pathophysiology of connective tissue diseases and cancer, and facilitating the design of novel strategies for tissue engineering.
Collapse
Affiliation(s)
- Iain Muntz
- Bionanoscience, TU Delft, Kavli Institute of Nanoscience Delft, Delft University of Technology, Van der Maasweg 9, Delft, Zuid-Holland, 2629 HC, NETHERLANDS
| | - Michele Fenu
- Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Zuid-Holland, 3000 CA, NETHERLANDS
| | - Gerjo J V M van Osch
- Orthopaedics; Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Zuid-Holland, 3000 CA, NETHERLANDS
| | - Gijsje Koenderink
- Bionanoscience, TU Delft, Kavli Institute of Nanoscience Delft, Delft University of Technology, Van der Maasweg 9, Delft, Zuid-Holland, 2629 HZ, NETHERLANDS
| |
Collapse
|
34
|
Zhang Y, Zhang Y, Liang H, Zhuo Z, Fan P, Chen Y, Zhang Z, Zhang W. Serum N-terminal DDR1: A Novel Diagnostic Marker of Liver Fibrosis Severity. J Clin Transl Hepatol 2021; 9:702-710. [PMID: 34722185 PMCID: PMC8516844 DOI: 10.14218/jcth.2021.00024] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/11/2021] [Accepted: 04/05/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND AIMS The expression of discoidin domain receptor 1 (DDR1) is commonly up-regulated and undergoes collagen-induced ectodomain (N-terminal) shedding during the progression of liver fibrosis. This study aimed to evaluate the clinical utility of N-terminal DDR1 as a diagnostic biomarker for liver fibrosis. METHODS N-terminal DDR1 shedding was evaluated using cell lines, liver fibrosis mouse models, clinical data of 298 patients collected from February 2019 to June 2020. The clinical data were divided into test and validation cohorts to evaluate the diagnostic performance of serum N-terminal DDR1. RESULTS Time- and dosage-dependent N-terminal DDR1 shedding stimulated by collagen I was observed in a hepatocyte cell line model. The type I collagen deposition and serum N-terminal DDR1 levels concurrently increased in the development of liver fibrosis in mouse models. Clinical data demonstrated a significant diagnostic power of serum N-terminal DDR1 levels as an accurate biomarker of liver fibrosis and cirrhosis. The diagnostic performance was further increased when applying N-DDR1/albumin ratio, achieving area under the curve of 0.790, 0.802, 0.879, and 0.865 for detecting histological fibrosis stages F ≥2, F ≥3, F 4 with liver biopsy as a reference method, and cirrhosis according to imaging techniques, respectively. With a cut-off of 55.6, a sensitivity, specificity, positive predictive value, and negative predictive value of 82.7%,76.6%, 67.4%, and 88.3% were achieved for the detection of cirrhosis. CONCLUSIONS Serum N-terminal DDR1 appears to be a novel diagnostic marker for liver fibrosis.
Collapse
Affiliation(s)
- Yuxin Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yujie Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zeng Zhuo
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Pan Fan
- Department of Surgery, University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yifa Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhanguo Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Correspondence to: Zhanguo Zhang and Wanguang Zhang, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei 430030, China. Tel: +86-2783665213, Fax: +86-27-83662640, E-mail: (ZZ) and (WZ)
| | - Wanguang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Correspondence to: Zhanguo Zhang and Wanguang Zhang, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei 430030, China. Tel: +86-2783665213, Fax: +86-27-83662640, E-mail: (ZZ) and (WZ)
| |
Collapse
|
35
|
Ma R, Xie X, Zhao L, Wu Y, Wang J. Discoidin domain receptors (DDRs): Potential implications in periodontitis. J Cell Physiol 2021; 237:189-198. [PMID: 34431091 DOI: 10.1002/jcp.30560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/01/2021] [Accepted: 08/09/2021] [Indexed: 02/05/2023]
Abstract
Periodontitis is a chronic inflammatory disease leading to the destruction of periodontal tissues associated with high prevalence and significant economic burden. As special collagen-binding tyrosine kinase receptors, the discoidin domain receptors (DDRs) can control cell migration, adhesion, proliferation, and extracellular matrix remodeling. DDRs are constitutively expressed and widely distributed in periodontal tissues which are rich in collagen. Ddr1/2 knockout mice showed significant periodontal defects including connective tissue destruction, alveolar bone loss, and even tooth loss. It has been demonstrated that bone homeostasis, inflammation, matrix metalloproteinases, and autophagy are crucial characteristics involved in the pathogenesis of periodontitis. Of note, DDRs have been reported to participate in the above pathophysiological processes, implicating the potential roles of DDRs in periodontitis. In this review article, we aim to illustrate the possible roles of DDRs in periodontitis in an attempt to explore their potential value as therapeutic targets for periodontitis.
Collapse
Affiliation(s)
- Rui Ma
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xudong Xie
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lei Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yafei Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
36
|
The Yin and Yang of Discoidin Domain Receptors (DDRs): Implications in Tumor Growth and Metastasis Development. Cancers (Basel) 2021; 13:cancers13071725. [PMID: 33917302 PMCID: PMC8038660 DOI: 10.3390/cancers13071725] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary The tumor microenvironment plays an important role in tumor development and metastasis. Collagens are major components of the extracellular matrix and can influence tumor development and metastasis by activating discoidin domain receptors (DDRs). This work shows the different roles of DDRs in various cancers and highlights the complexity of anti-DDR therapies in cancer treatment. Abstract The tumor microenvironment is a complex structure composed of the extracellular matrix (ECM) and nontumoral cells (notably cancer-associated fibroblasts (CAFs) and immune cells). Collagens are the main components of the ECM and they are extensively remodeled during tumor progression. Some collagens are ligands for the discoidin domain receptor tyrosine kinases, DDR1 and DDR2. DDRs are involved in different stages of tumor development and metastasis formation. In this review, we present the different roles of DDRs in these processes and discuss controversial findings. We conclude by describing emerging DDR inhibitory strategies, which could be used as new alternatives for the treatment of patients.
Collapse
|
37
|
Ostrowska-Podhorodecka Z, Ding I, Lee W, Tanic J, Abbasi S, Arora PD, Liu RS, Patteson AE, Janmey PA, McCulloch CA. Vimentin tunes cell migration on collagen by controlling β1 integrin activation and clustering. J Cell Sci 2021; 134:jcs.254359. [PMID: 33558312 DOI: 10.1242/jcs.254359] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/20/2021] [Indexed: 12/17/2022] Open
Abstract
Vimentin is a structural protein that is required for mesenchymal cell migration and directly interacts with actin, β1 integrin and paxillin. We examined how these interactions enable vimentin to regulate cell migration on collagen. In fibroblasts, depletion of vimentin increased talin-dependent activation of β1 integrin by more than 2-fold. Loss of vimentin was associated with reduction of β1 integrin clustering by 50% and inhibition of paxillin recruitment to focal adhesions by more than 60%, which was restored by vimentin expression. This reduction of paxillin was associated with 65% lower Cdc42 activation, a 60% reduction of cell extension formation and a greater than 35% decrease in cell migration on collagen. The activation of PAK1, a downstream effector of Cdc42, was required for vimentin phosphorylation and filament maturation. We propose that vimentin tunes cell migration through collagen by acting as an adaptor protein for focal adhesion proteins, thereby regulating β1 integrin activation, resulting in well-organized, mature integrin clusters.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | - Isabel Ding
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Wilson Lee
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Jelena Tanic
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Sevil Abbasi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Pamma D Arora
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Richard S Liu
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Alison E Patteson
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104-6393, USA.,Physics Department, Syracuse University, Syracuse, NY 13244, USA
| | - Paul A Janmey
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104-6393, USA
| | | |
Collapse
|
38
|
Musiime M, Chang J, Hansen U, Kadler KE, Zeltz C, Gullberg D. Collagen Assembly at the Cell Surface: Dogmas Revisited. Cells 2021; 10:662. [PMID: 33809734 PMCID: PMC8002325 DOI: 10.3390/cells10030662] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/12/2022] Open
Abstract
With the increased awareness about the importance of the composition, organization, and stiffness of the extracellular matrix (ECM) for tissue homeostasis, there is a renewed need to understand the details of how cells recognize, assemble and remodel the ECM during dynamic tissue reorganization events. Fibronectin (FN) and fibrillar collagens are major proteins in the ECM of interstitial matrices. Whereas FN is abundant in cell culture studies, it is often only transiently expressed in the acute phase of wound healing and tissue regeneration, by contrast fibrillar collagens form a persistent robust scaffold in healing and regenerating tissues. Historically fibrillar collagens in interstitial matrices were seen merely as structural building blocks. Cell anchorage to the collagen matrix was thought to be indirect and occurring via proteins like FN and cell surface-mediated collagen fibrillogenesis was believed to require a FN matrix. The isolation of four collagen-binding integrins have challenged this dogma, and we now know that cells anchor directly to monomeric forms of fibrillar collagens via the α1β1, α2β1, α10β1 and α11β1 integrins. The binding of these integrins to the mature fibrous collagen matrices is more controversial and depends on availability of integrin-binding sites. With increased awareness about the importance of characterizing the total integrin repertoire on cells, including the integrin collagen receptors, the idea of an absolute dependence on FN for cell-mediated collagen fibrillogenesis needs to be re-evaluated. We will summarize data suggesting that collagen-binding integrins in vitro and in vivo are perfectly well suited for nucleating and supporting collagen fibrillogenesis, independent of FN.
Collapse
Affiliation(s)
- Moses Musiime
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway; (M.M.); (C.Z.)
| | - Joan Chang
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (J.C.); (K.E.K.)
| | - Uwe Hansen
- Institute for Musculoskeletal Medicine, University Hospital of Münster, 48149 Münster, Germany;
| | - Karl E. Kadler
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (J.C.); (K.E.K.)
| | - Cédric Zeltz
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway; (M.M.); (C.Z.)
| | - Donald Gullberg
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway; (M.M.); (C.Z.)
| |
Collapse
|
39
|
Abstract
Human hypertrophic scars are the result of imperfect healing of skin, which is particularly evident from the scars developing after severe burns. In contrast, mouse and rat full-thickness skin wounds heal normally without forming visible scar tissue, which reduces the suitability of rodent models for the study of skin scarring. We here provide a simple procedure to splint the edges of full-thickness rodent skin with a sutured plastic frame that prevents wound closure by granulation tissue contraction. The resulting mechanical tension in the wound bed and the lack of neo-epithelium amplify myofibroblast formation and generate hypertrophic features, not unlike those of human skin. In addition to producing scar tissue, the splint provides a reservoir that can be used for the delivery of cellular and acellular wound treatment regimen. Despite being simple and almost historical, wound splinting is a robust and reliable model to study myofibroblast biology.
Collapse
Affiliation(s)
- Dong Ok Son
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
40
|
Kuroda J, Itabashi T, Iwane AH, Aramaki T, Kondo S. The Physical Role of Mesenchymal Cells Driven by the Actin Cytoskeleton Is Essential for the Orientation of Collagen Fibrils in Zebrafish Fins. Front Cell Dev Biol 2020; 8:580520. [PMID: 33154970 PMCID: PMC7591588 DOI: 10.3389/fcell.2020.580520] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022] Open
Abstract
Fibrous collagen imparts physical strength and flexibility to tissues by forming huge complexes. The density and orientation of collagen fibers must be correctly specified for the optimal physical property of the collagen complex. However, little is known about its underlying cellular mechanisms. Actinotrichia are collagen fibers aligned at the fin-tip of bony fish and are easily visible under the microscope due to their thick, linear structure. We used the actinotrichia as a model system to investigate how cells manipulate collagen fibers. The 3D image obtained by focused ion beam scanning electron microscopy (FIB-SEM) showed that the pseudopodia of mesenchymal cells encircle the multiple actinotrichia. We then co-incubated the mesenchymal cells and actinotrichia in vitro, and time-lapse analysis revealed how cells use pseudopods to align collagen fiber orientation. This in vitro behavior is dependent on actin polymerization in mesenchymal cells. Inhibition of actin polymerization in mesenchymal cells results in mis-orientation of actinotrichia in the fin. These results reveal how mesenchymal cells are involved in fin formation and have important implications for the physical interaction between cells and collagen fibers.
Collapse
Affiliation(s)
- Junpei Kuroda
- Graduate School of Frontier Bioscience, Osaka University, Suita, Japan
- RIKEN Center for Biosystems Dynamics Research, Higashi-Hiroshima, Japan
| | - Takeshi Itabashi
- RIKEN Center for Biosystems Dynamics Research, Higashi-Hiroshima, Japan
| | - Atsuko H. Iwane
- RIKEN Center for Biosystems Dynamics Research, Higashi-Hiroshima, Japan
| | - Toshihiro Aramaki
- Graduate School of Frontier Bioscience, Osaka University, Suita, Japan
| | - Shigeru Kondo
- Graduate School of Frontier Bioscience, Osaka University, Suita, Japan
| |
Collapse
|
41
|
Xu R, Zhou X, Wang S, Trinkle C. Tumor organoid models in precision medicine and investigating cancer-stromal interactions. Pharmacol Ther 2020; 218:107668. [PMID: 32853629 DOI: 10.1016/j.pharmthera.2020.107668] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023]
Abstract
Tumor development and progression require chemical and mechanical cues derived from cellular and non-cellular components in the tumor microenvironment, including the extracellular matrix (ECM), cancer-associated fibroblasts (CAFs), endothelial cells, and immune cells. Therefore, it is crucial to develop tissue culture models that can mimic in vivo cancer cell-ECM and cancer-stromal cell interactions. Three-dimensional (3D) tumor culture models have been widely utilized to study cancer development and progression. A recent advance in 3D culture is the development of patient-derived tumor organoid (PDO) models from primary human cancer tissue. PDOs maintain the heterogeneity of the primary tumor, which makes them more relevant for identifying therapeutic targets and verifying drug response. Other significant advances include development of 3D co-culture assays to investigate cell-cell interactions and tissue/organ morphogenesis, and microfluidic technology that can be integrated into 3D culture to mimic vasculature and blood flow. These advances offer spatial and temporal insights into cancer cell-stromal interactions and represent novel techniques to study tumor progression and drug response. Here, we summarize the recent progress in 3D culture and tumor organoid models, and discuss future directions and the potential of utilizing these models to study cancer-stromal interactions and direct personalized treatment.
Collapse
Affiliation(s)
- Ren Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40508, USA.
| | - Xiaotao Zhou
- Markey Cancer Center, University of Kentucky, Lexington, KY 40508, USA
| | - Shike Wang
- Markey Cancer Center, University of Kentucky, Lexington, KY 40508, USA
| | - Christine Trinkle
- Department of Mechanical Engineering, University of Kentucky, Lexington, KY, 40506, USA
| |
Collapse
|
42
|
Coelho NM, Wang A, Petrovic P, Wang Y, Lee W, McCulloch CA. MRIP Regulates the Myosin IIA Activity and DDR1 Function to Enable Collagen Tractional Remodeling. Cells 2020; 9:cells9071672. [PMID: 32664526 PMCID: PMC7407560 DOI: 10.3390/cells9071672] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/18/2022] Open
Abstract
DDR1 is a collagen adhesion-mechanoreceptor expressed in fibrotic lesions. DDR1 mediates non-muscle myosin IIA (NMIIA)-dependent collagen remodeling. We discovered that the myosin phosphatase Rho-interacting protein (MRIP), is enriched in DDR1-NMIIA adhesions on collagen. MRIP regulates RhoA- and myosin phosphatase-dependent myosin activity. We hypothesized that MRIP regulates DDR1-NMIIA interactions to enable cell migration and collagen tractional remodeling. After deletion of MRIP in β1-integrin null cells expressing DDR1, in vitro wound closure, collagen realignment, and contraction were reduced. Cells expressing DDR1 and MRIP formed larger and more abundant DDR1 clusters on collagen than cells cultured on fibronectin or cells expressing DDR1 but null for MRIP or cells expressing a non-activating DDR1 mutant. Deletion of MRIP reduced DDR1 autophosphorylation and blocked myosin light chain-dependent contraction. Deletion of MRIP did not disrupt the association of DDR1 with NMIIA. We conclude that MRIP regulates NMIIA-dependent DDR1 cluster growth and activation. Accordingly, MRIP may provide a novel drug target for dysfunctional DDR1-related collagen tractional remodeling in fibrosis.
Collapse
|
43
|
Wang Q, Notay K, Downey GP, McCulloch CA. The Leucine-Rich Repeat Region of CARMIL1 Regulates IL-1-Mediated ERK Activation, MMP Expression, and Collagen Degradation. Cell Rep 2020; 31:107781. [PMID: 32610117 PMCID: PMC8713033 DOI: 10.1016/j.celrep.2020.107781] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/29/2020] [Accepted: 05/27/2020] [Indexed: 01/08/2023] Open
Abstract
CARMILs are large, multidomain, membrane-associated proteins that regulate actin assembly and Rho-family GTPases, but their role in inflammatory signaling is not defined. Tandem mass tag mass spectrometry indicated that, in fibroblasts, CARMIL1 associates with interleukin (IL)-1 signaling molecules. Immunoprecipitation of cells transfected with CARMIL1 mutants showed that the leucine-rich repeat (LRR) region of CARMIL1 associates with IL-1 receptor type 1 (IL-1R1) and IL-1 receptor-associated kinase (IRAK). Knockout of CARMIL1 by CRISPR-Cas9 reduced IL-1-induced ERK activation by 72% and MMP3 expression by 40%. Compared with CARMIL1 wild-type (WT), cells expressing mutant CARMIL1 lacking its LRR domain exhibited 45% lower ERK activation and 40% lower MMP3 expression. In fibroblasts transduced with a cell-permeable, TAT CARMIL1 peptide that competed with IL-1R1 and IRAK binding to the LRR of CARMIL1, collagen degradation was reduced by 43%. As the LRR of CARMIL1 evidently regulates IL-1 signaling, CARMIL1 could become a target for anti-inflammatory drug development.
Collapse
Affiliation(s)
- Qin Wang
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada.
| | - Karambir Notay
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada.
| | - Gregory P Downey
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, National Jewish Health, Denver, CO 80206, USA.
| | - Christopher A McCulloch
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada; Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, National Jewish Health, Denver, CO 80206, USA.
| |
Collapse
|
44
|
El-Hattab MY, Nagumo Y, Gourronc FA, Klingelhutz AJ, Ankrum JA, Sander EA. Human Adipocyte Conditioned Medium Promotes In Vitro Fibroblast Conversion to Myofibroblasts. Sci Rep 2020; 10:10286. [PMID: 32581231 PMCID: PMC7314785 DOI: 10.1038/s41598-020-67175-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
Adipocytes and adipose tissue derived cells have been investigated for their potential to contribute to the wound healing process. However, the details of how these cells interact with other essential cell types, such as myofibroblasts/fibroblasts, remain unclear. Using a novel in-vitro 3D human adipocyte/pre-adipocyte spheroid model, we investigated whether adipocytes and their precursors (pre-adipocytes) secrete factors that affect human dermal fibroblast behavior. We found that both adipocyte and pre-adipocyte conditioned medium induced the migration of fibroblasts, but only adipocyte conditioned medium induced fibroblast differentiation into a highly contractile, collagen producing myofibroblast phenotype. Furthermore, adipocyte mediated myofibroblast induction occurred through a TGF-β independent mechanism. Our findings contribute to a better understanding on the involvement of adipose tissue in wound healing, and may help to uncover and develop fat-related wound healing treatments.
Collapse
Affiliation(s)
- Mariam Y El-Hattab
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA, USA
| | - Yoshiaki Nagumo
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA, USA
- Department of Plastic Surgery, Kindai University, Faculty of Medicine, Higashiosaka, Osaka, Japan
| | - Francoise A Gourronc
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Aloysius J Klingelhutz
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA
| | - James A Ankrum
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA, USA.
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA.
| | - Edward A Sander
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA, USA.
- Department of Orthopedics and Rehabilitation, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
45
|
Ngai D, Lino M, Rothenberg KE, Simmons CA, Fernandez-Gonzalez R, Bendeck MP. DDR1 (Discoidin Domain Receptor-1)-RhoA (Ras Homolog Family Member A) Axis Senses Matrix Stiffness to Promote Vascular Calcification. Arterioscler Thromb Vasc Biol 2020; 40:1763-1776. [PMID: 32493168 PMCID: PMC7310304 DOI: 10.1161/atvbaha.120.314697] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Supplemental Digital Content is available in the text. Objective: Vascular calcification is a pathology characterized by arterial mineralization, which is a common late-term complication of atherosclerosis that independently increases the risk of adverse cardiovascular events by fourfold. A major source of calcifying cells is transdifferentiating vascular smooth muscle cells (VSMCs). Previous studies showed that deletion of the collagen-binding receptor, DDR1 (discoidin domain receptor-1), attenuated VSMC calcification. Increased matrix stiffness drives osteogenesis, and DDR1 has been implicated in stiffness sensing in other cell types; however, the role of DDR1 as a mechanosensor in VSMCs has not been investigated. Here, we test the hypothesis that DDR1 senses increased matrix stiffness and promotes VSMC transdifferentiation and calcification. Approach and Results: Primary VSMCs isolated from Ddr1+/+ (wild-type) and Ddr1−/− (knockout) mice were studied on collagen-I–coated silicon substrates of varying stiffness, culturing in normal or calcifying medium. DDR1 expression and phosphorylation increased with increasing stiffness, as did in vitro calcification, nuclear localization of Runx2 (Runt-related transcription factor 2), and expression of other osteochondrocytic markers. By contrast, DDR1 deficient VSMCs were not responsive to stiffness and did not undergo transdifferentiation. DDR1 regulated stress fiber formation and RhoA (ras homolog family member A) activation through the RhoGEF (rho guanine nucleotide exchange factor), Vav2. Inhibition of actomyosin contractility reduced Runx2 activation and attenuated in vitro calcification in wild-type VSMCs. Finally, a novel positive feedforward loop was uncovered between DDR1 and actomyosin contractility, important in regulating DDR1 expression, clustering, and activation. Conclusions: This study provides mechanistic insights into DDR1 mechanosignaling and shows that DDR1 activity and actomyosin contractility are interdependent in mediating stiffness-dependent increases in VSMC calcification.
Collapse
Affiliation(s)
- David Ngai
- From the Department of Laboratory Medicine and Pathobiology (D.N., M.L., M.P.B.), University of Toronto, Canada.,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research (D.N. M.L., K.E.R., C.A.S., R.F.-G., M.P.B.), University of Toronto, Canada
| | - Marsel Lino
- From the Department of Laboratory Medicine and Pathobiology (D.N., M.L., M.P.B.), University of Toronto, Canada.,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research (D.N. M.L., K.E.R., C.A.S., R.F.-G., M.P.B.), University of Toronto, Canada
| | - Katheryn E Rothenberg
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research (D.N. M.L., K.E.R., C.A.S., R.F.-G., M.P.B.), University of Toronto, Canada.,Institute of Biomaterials and Biomedical Engineering (K.E.R., C.A.S., R.F.-G.), University of Toronto, Canada
| | - Craig A Simmons
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research (D.N. M.L., K.E.R., C.A.S., R.F.-G., M.P.B.), University of Toronto, Canada.,Institute of Biomaterials and Biomedical Engineering (K.E.R., C.A.S., R.F.-G.), University of Toronto, Canada.,Department of Mechanical and Industrial Engineering (C.A.S.), University of Toronto, Canada
| | - Rodrigo Fernandez-Gonzalez
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research (D.N. M.L., K.E.R., C.A.S., R.F.-G., M.P.B.), University of Toronto, Canada.,Institute of Biomaterials and Biomedical Engineering (K.E.R., C.A.S., R.F.-G.), University of Toronto, Canada.,Department of Cell and Systems Biology (R.F.-G.), University of Toronto, Canada
| | - Michelle P Bendeck
- From the Department of Laboratory Medicine and Pathobiology (D.N., M.L., M.P.B.), University of Toronto, Canada.,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research (D.N. M.L., K.E.R., C.A.S., R.F.-G., M.P.B.), University of Toronto, Canada
| |
Collapse
|
46
|
Discoidin domain receptor 1-deletion ameliorates fibrosis and promotes adipose tissue beiging, brown fat activity, and increased metabolic rate in a mouse model of cardiometabolic disease. Mol Metab 2020; 39:101006. [PMID: 32360427 PMCID: PMC7242876 DOI: 10.1016/j.molmet.2020.101006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/10/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
Objective Discoidin domain receptor 1 (DDR1) is a collagen binding receptor tyrosine kinase implicated in atherosclerosis, fibrosis, and cancer. Our previous research showed that DDR1 could regulate smooth muscle cell trans-differentiation, fibrosis and calcification in the vascular system in cardiometabolic disease. This spectrum of activity led us to question whether DDR1 might also regulate adipose tissue fibrosis and remodeling. Methods We have used a diet-induced mouse model of cardiometabolic disease to determine whether DDR1 deletion impacts upon adipose tissue remodeling and metabolic dysfunction. Mice were fed a high fat diet (HFD) for 12 weeks, followed by assessment of glucose and insulin tolerance, respiration via indirect calorimetry, and brown fat activity by FDG-PET. Results Feeding HFD induced DDR1 expression in white adipose tissue, which correlated with adipose tissue expansion and fibrosis. Ddr1−/− mice fed an HFD had improved glucose tolerance, reduced body fat, and increased brown fat activity and energy expenditure compared to Ddr1+/+ littermate controls. HFD-fed DDR1−/− mice also had reduced fibrosis, smaller adipocytes with multilocular lipid droplets, and increased UCP-1 expression characteristic of beige fat formation in subcutaneous adipose tissue. In vitro, studying C3H10T1/2 cells stimulated to differentiate, DDR1 inhibition caused a shift from white to beige adipocyte differentiation, whereas DDR1 expression was increased with TGFβ-mediated pro-fibrotic differentiation. Conclusion This study is the first to identify a role for DDR1 as a driver of adipose tissue fibrosis and suppressor of beneficial beige fat formation. DDR1 deletion results in decreased obesity, and increased energy expenditure and brown fat activity. DDR1 expression was increased in adipose and correlated with obesity and fibrosis. DDR1 deletion increased UCP-1 expression in brown and white fat in vivo, and in mesenchymal cells in vitro. Invitro studies suggest that DDR1 suppresses UCP-1 and drives pro-fibrotic differentiation of mesenchymal cells.
Collapse
|
47
|
Lafitte M, Sirvent A, Roche S. Collagen Kinase Receptors as Potential Therapeutic Targets in Metastatic Colon Cancer. Front Oncol 2020; 10:125. [PMID: 32117772 PMCID: PMC7028753 DOI: 10.3389/fonc.2020.00125] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/23/2020] [Indexed: 12/19/2022] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of tumor-related death worldwide. While surgery can cure patients with early stage CRC, the 5-year survival rate is only 10% for patients with metastatic disease. Therefore, new anti-metastatic therapies are needed for this cancer. Metastatic spread defines the dissemination of cancer cells with tumor-initiating capacities from the primary tumor and their colonization of distinct organs, mainly the liver, for secondary tumor formation. Although the underlying mechanisms are not fully understood, components of the tumor microenvironment have gained strong interest. Among the known metastatic-promoting factors, collagens are extracellular matrix components that are deposited within the tumor, the tumor microenvironment, and at metastatic site(s), and are recognized to play essential roles during metastasis development. Here, we review recent findings on the metastatic role of the collagen receptors Discoidin Domain Receptors 1 and 2 (DDR1 and DDR2) in CRC and discuss the therapeutic value of targeting these receptor tyrosine kinases in this cancer.
Collapse
Affiliation(s)
| | | | - Serge Roche
- CRBM, CNRS, Univ. Montpellier, Montpellier, France
| |
Collapse
|
48
|
Wasinski B, Sohail A, Bonfil RD, Kim S, Saliganan A, Polin L, Bouhamdan M, Kim HRC, Prunotto M, Fridman R. Discoidin Domain Receptors, DDR1b and DDR2, Promote Tumour Growth within Collagen but DDR1b Suppresses Experimental Lung Metastasis in HT1080 Xenografts. Sci Rep 2020; 10:2309. [PMID: 32047176 PMCID: PMC7012844 DOI: 10.1038/s41598-020-59028-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/20/2020] [Indexed: 12/17/2022] Open
Abstract
The Discoidin Domain Receptors (DDRs) constitute a unique set of receptor tyrosine kinases that signal in response to collagen. Using an inducible expression system in human HT1080 fibrosarcoma cells, we investigated the role of DDR1b and DDR2 on primary tumour growth and experimental lung metastases. Neither DDR1b nor DDR2 expression altered tumour growth at the primary site. However, implantation of DDR1b- or DDR2-expressing HT1080 cells with collagen I significantly accelerated tumour growth rate, an effect that could not be observed with collagen I in the absence of DDR induction. Interestingly, DDR1b, but not DDR2, completely hindered the ability of HT1080 cells to form lung colonies after intravenous inoculation, suggesting a differential role for DDR1b in primary tumour growth and lung colonization. Analyses of tumour extracts revealed specific alterations in Hippo pathway core components, as a function of DDR and collagen expression, that were associated with stimulation of tumour growth by DDRs and collagen I. Collectively, these findings identified divergent effects of DDRs on primary tumour growth and experimental lung metastasis in the HT1080 xenograft model and highlight the critical role of fibrillar collagen and DDRs in supporting the growth of tumours thriving within a collagen-rich stroma.
Collapse
Affiliation(s)
- Benjamin Wasinski
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Anjum Sohail
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - R Daniel Bonfil
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA.,Department of Urology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA.,Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA.,Department of Pathology, College of Medical Sciences and Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, 33328-2018, USA
| | - Seongho Kim
- Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Allen Saliganan
- Department of Urology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Lisa Polin
- Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Mohamad Bouhamdan
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Hyeong-Reh C Kim
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA.,Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Marco Prunotto
- Hoffmann-La Roche, Basel, Switzerland.,School of Pharmaceutical Sciences, Geneva, Switzerland
| | - Rafael Fridman
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA. .,Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA.
| |
Collapse
|
49
|
DDR1 autophosphorylation is a result of aggregation into dense clusters. Sci Rep 2019; 9:17104. [PMID: 31745115 PMCID: PMC6863838 DOI: 10.1038/s41598-019-53176-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/25/2019] [Indexed: 12/11/2022] Open
Abstract
The collagen receptor DDR1 is a receptor tyrosine kinase that promotes progression of a wide range of human disorders. Little is known about how ligand binding triggers DDR1 kinase activity. We previously reported that collagen induces DDR1 activation through lateral dimer association and phosphorylation between dimers, a process that requires specific transmembrane association. Here we demonstrate ligand-induced DDR1 clustering by widefield and super-resolution imaging and provide evidence for a mechanism whereby DDR1 kinase activity is determined by its molecular density. Ligand binding resulted in initial DDR1 reorganisation into morphologically distinct clusters with unphosphorylated DDR1. Further compaction over time led to clusters with highly aggregated and phosphorylated DDR1. Ligand-induced DDR1 clustering was abolished by transmembrane mutations but did not require kinase activity. Our results significantly advance our understanding of the molecular events underpinning ligand-induced DDR1 kinase activity and provide an explanation for the unusually slow DDR1 activation kinetics.
Collapse
|
50
|
Chavez MB, Kolli TN, Tan MH, Zachariadou C, Wang C, Embree MC, Lira Dos Santos EJ, Nociti FH, Wang Y, Tatakis DN, Agarwal G, Foster BL. Loss of Discoidin Domain Receptor 1 Predisposes Mice to Periodontal Breakdown. J Dent Res 2019; 98:1521-1531. [PMID: 31610730 DOI: 10.1177/0022034519881136] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The discoidin domain receptors, DDR1 and DDR2, are nonintegrin collagen receptors and tyrosine kinases. DDRs regulate cell functions, and their extracellular domains affect collagen fibrillogenesis and mineralization. Based on the collagenous nature of dentoalveolar tissues, we hypothesized that DDR1 plays an important role in dentoalveolar development and function. Radiography, micro-computed tomography (micro-CT), histology, histomorphometry, in situ hybridization (ISH), immunohistochemistry (IHC), and transmission electron microscopy (TEM) were used to analyze Ddr1 knockout (Ddr1-/-) mice and wild-type (WT) controls at 1, 2, and 9 mo, and ISH and quantitative polymerase chain reaction (qPCR) were employed to assess Ddr1/DDR1 messenger RNA expression in mouse and human tissues. Radiographic images showed normal molars but abnormal mandibular condyles, as well as alveolar bone loss in Ddr1-/- mice versus WT controls at 9 mo. Histological, histomorphometric, micro-CT, and TEM analyses indicated no differences in enamel or dentin Ddr1-/- versus WT molars. Total volumes (TVs) and bone volumes (BVs) of subchondral and ramus bone of Ddr1-/- versus WT condyles were increased and bone volume fraction (BV/TV) was reduced at 1 and 9 mo. There were no differences in alveolar bone volume at 1 mo, but at 9 mo, severe periodontal defects and significant alveolar bone loss (14%; P < 0.0001) were evident in Ddr1-/- versus WT mandibles. Histology, ISH, and IHC revealed disrupted junctional epithelium, connective tissue destruction, bacterial invasion, increased neutrophil infiltration, upregulation of cytokines including macrophage colony-stimulating factor, and 3-fold increased osteoclast numbers (P < 0.05) in Ddr1-/- versus WT periodontia at 9 mo. In normal mouse tissues, ISH and qPCR revealed Ddr1 expression in basal cell layers of the oral epithelia and in immune cells. We confirmed a similar expression pattern in human oral epithelium by ISH and qPCR. We propose that DDR1 plays an important role in periodontal homeostasis and that absence of DDR1 predisposes mice to periodontal breakdown.
Collapse
Affiliation(s)
- M B Chavez
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - T N Kolli
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - M H Tan
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - C Zachariadou
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - C Wang
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - M C Embree
- TMJ Biology and Regenerative Medicine Laboratory, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - E J Lira Dos Santos
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA.,Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas-UNICAMP, Piracicaba, SP, Brazil
| | - F H Nociti
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas-UNICAMP, Piracicaba, SP, Brazil
| | - Y Wang
- Division of Periodontology, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - D N Tatakis
- Division of Periodontology, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - G Agarwal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - B L Foster
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| |
Collapse
|