1
|
Moreno SE, Enwerem-Lackland I, Dreaden K, Massee M, Koob TJ, Harper JR. Human amniotic membrane modulates collagen production and deposition in vitro. Sci Rep 2024; 14:15998. [PMID: 38987293 PMCID: PMC11237048 DOI: 10.1038/s41598-024-64364-2] [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: 11/10/2023] [Accepted: 06/07/2024] [Indexed: 07/12/2024] Open
Abstract
Pathological fibrosis is a significant complication of surgical procedures resulting from the accumulation of excess collagen at the site of repair which can compromise the tissue architecture and severely impede the function of the affected tissue. Few prophylactic treatments exist to counteract this process; however, the use of amniotic membrane allografts has demonstrated promising clinical outcomes. This study aimed to identify the underlying mechanism of action by utilizing relevant models that accurately represent the pathophysiology of the disease state. This study employed a pro-fibrotic in vitro system using TGFβ1 stimulation and macromolecular crowding techniques to evaluate the mechanism by which amniotic membrane allografts regulate collagen biosynthesis and deposition. Following treatment with dehydrated human amnion chorion membrane (DHACM), subsequent RNA sequencing and functional enrichment with Reactome pathway analysis indicated that amniotic membranes are indeed capable of regulating genes associated with the composition and function of the extracellular matrix. Furthermore, macromolecular crowding was used in vitro to expand the evaluation to include both the effects of DHACM and a lyophilized human amnion/chorion membrane (LHACM). DHACM and LHACM regulate the TGFβ pathway and myofibroblast differentiation. Additionally, both DHACM and LHACM modulate the production, secretion, and deposition of collagen type I, a primary target for pathological fibrosis. These observations support the hypothesis that amniotic membranes may interrupt pathological fibrosis by regulating collagen biosynthesis and associated pathways.
Collapse
Affiliation(s)
- Sarah E Moreno
- MIMEDX Group, Inc., 1775 West Oak Commons Court NE, Marietta, GA, 30062, USA
| | | | | | - Michelle Massee
- MIMEDX Group, Inc., 1775 West Oak Commons Court NE, Marietta, GA, 30062, USA.
| | - Thomas J Koob
- MIMEDX Group, Inc., 1775 West Oak Commons Court NE, Marietta, GA, 30062, USA
| | - John R Harper
- MIMEDX Group, Inc., 1775 West Oak Commons Court NE, Marietta, GA, 30062, USA
| |
Collapse
|
2
|
Wang Y, Chen Y, Wu J, Shi X. BMP1 Promotes Keloid by Inducing Fibroblast Inflammation and Fibrogenesis. J Cell Biochem 2024; 125:e30609. [PMID: 38860429 DOI: 10.1002/jcb.30609] [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: 12/15/2023] [Revised: 05/11/2024] [Accepted: 05/24/2024] [Indexed: 06/12/2024]
Abstract
Keloid is a typical fibrotic and inflammatory skin disease with unclear mechanisms and few therapeutic targets. In this study, we found that BMP1 was significantly increased in a collagen high-expressing subtype of fibroblast by reanalyzing a public single-cell RNA-sequence data set of keloid. The number of BMP1-positive fibroblast cells was increased in keloid fibrotic loci. Increased levels of BMP1 were further validated in the skin tissues and fibroblasts from keloid patients. Additionally, a positive correlation between BMP1 and the Keloid Area and Severity Index was found in keloid patients. In vitro analysis revealed collagen production, the phosphorylation levels of p65, and the IL-1β secretion decreased in BMP1 interfered keloid fibroblasts. Besides, the knockdown of BMP1 inhibited the growth and migration of keloid fibroblast cells. Mechanistically, BMP1 inhibition downregulated the noncanonical TGF-β pathways, including p-p38 and p-ERK1/2 signaling. Furthermore, we found the delivery of BMP1 siRNAs could significantly alleviate keloid in human keloid-bearing nude mice. Collectively, our results indicated that BMP1 exhibited various pathogenic effects on keloids as promoting cell proliferation, migration, inflammation, and ECM deposition of fibroblast cells by regulating the noncanonical TGF-β/p38 MAPK, and TGF-β/ERK pathways. BMP1-lowing strategies may appear as a potential new therapeutic target for keloid.
Collapse
Affiliation(s)
- Yi Wang
- Department of Plastic and Burns Surgery, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Yahui Chen
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Jinfeng Wu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiangguang Shi
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Patricelli C, Lehmann P, Oxford JT, Pu X. Doxorubicin-Induced Modulation of TGF-β Signaling Cascade in Mouse Fibroblasts: Insights into Cardiotoxicity Mechanisms. RESEARCH SQUARE 2023:rs.3.rs-3186393. [PMID: 37546862 PMCID: PMC10402200 DOI: 10.21203/rs.3.rs-3186393/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Doxorubicin (DOX)-induced cardiotoxicity has been widely observed, yet the specific impact on cardiac fibroblasts is not fully understood. Additionally, the modulation of the transforming growth factor beta (TGF-β) signaling pathway by DOX remains to be fully elucidated. This study investigated DOX's ability to modulate the expression of genes and proteins involved in the TGF-β signaling cascade in mouse fibroblasts from two sources by assessing the impact of DOX treatment on TGF-β inducible expression of pivotal genes and proteins within fibroblasts. Mouse embryonic fibroblasts (NIH3T3) and mouse primary cardiac fibroblasts (CFs) were treated with DOX in the presence of TGF-β1 to assess changes in protein levels by western blot and changes in mRNA levels by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). Our results revealed a dose-dependent reduction in cellular communication network factor 2 (CCN2) protein levels upon DOX treatment in both NIH3T3 and CFs. Moreover, we observed that DOX inhibited the TGF-β1 induced expression of BMP1 in NIH3T3 cells, while BMP1 levels remained high in CFs, and that TGF-β1 induces the phosphorylation of SMAD2 in both NIH3T3 cells and CFs. While DOX treatment diminished the extent of phosphorylation, the reduction did not reach statistical significance. DOX also inhibited the TGF-β1 induced expression of COL1 in NIH3T3 cells and CFs. Finally, DOX inhibited the TGF-β1 induced expression of Atf4 and increased the expression of Cdkn1a, Id1, Id2, Runx1, Tgfb1, Inhba, Thbs1, Bmp1, and Stat1 in NIH3T3 cells but not CFs, indicating the potential for cell-specific responses to DOX and its modulation of the TGF-β signaling pathway. Understanding the underlying mechanisms of the ability of DOX to modulate gene expression and signaling pathways in fibroblasts holds promise for future development of targeted therapeutic strategies to mitigate DOX-induced cardiotoxicity specifically affecting CFs.
Collapse
|
4
|
Yifan H, Peng G, Tao Q, Bo C, Tao X, Jiang Y, Qian W, Zhenqi Y, Tao J, Jin F, Shujie Z, Wei Z, Jian C, Guoyong Y. Delayed inhibition of collagen deposition by targeting bone morphogenetic protein 1 promotes recovery after spinal cord injury. Matrix Biol 2023; 118:69-91. [PMID: 36918086 DOI: 10.1016/j.matbio.2023.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 03/13/2023]
Abstract
Fibrotic scars appear after spinal cord injury (SCI) and are mainly composed of fibroblasts and excess extracellular matrix (ECM), including different types of collagen. The temporal and spatial distribution and role of excess collagens and ECM after SCI are not yet fully understood. Here, we identified that the procollagen type I C-terminal propeptide (PICP), a marker of collagen type I deposition, and bone morphogenetic protein 1 (BMP1), a secreted procollagen c-proteinase (PCP) for type I collagen maturation, were significantly elevatedin cerebrospinal fluid of patients with SCI compared with healthy controls, and were associated with spinal cord compression and neurological symptoms. We revealed the deposition of type I collagen in the area damaged by SCI in mice and confirmed that BMP1 was the only expressed PCP and induced collagen deposition. Furthermore, transforming growth factor-β (TGF-β), tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) can activate the expression of BMP1. However, inhibition of BMP1 at the acute phase eliminated fibrotic scars in the damaged area and inhibited activation and enrichment of astrocytes, which made the damage difficult to repair and increased hematoma. Unexpectedly, knockdown of Bmp1 by adeno-associated virus or the inhibition of BMP1 biological function by specific inhibitors and monoclonal antibodies at different time points after injury led to distinct therapeutic effects. Only delayed inhibition of BMP1 improved axonal regeneration and myelin repair at the subacute stage post-injury, and led to the recovery of motor function, suggesting that scarring had a dual effect. Early inhibition of the scarring was not conducive to limiting inflammation, while excessive scar formation inhibited the growth of axons. After SCI, the collagen deposition indicators increased in both human cerebrospinal fluid and mouse spinal cord. Therefore, suppression of BMP1 during the subacute phase improves nerve function after SCI and is a potential target for scar reduction.
Collapse
Affiliation(s)
- Huang Yifan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Gao Peng
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Qin Tao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Chu Bo
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Xu Tao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Yi Jiang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Wang Qian
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Yang Zhenqi
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Jiang Tao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Fan Jin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Zhao Shujie
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China.
| | - Zhou Wei
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China.
| | - Chen Jian
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China.
| | - Yin Guoyong
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
5
|
Mechanochemistry of collagen. Acta Biomater 2023; 163:50-62. [PMID: 36669548 DOI: 10.1016/j.actbio.2023.01.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 01/02/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023]
Abstract
The collagen molecular family is the result of nearly one billion years of evolution. It is a unique family of proteins, the majority of which provide general mechanical support to biological tissues. Fibril forming collagens are the most abundant collagens in vertebrate animals and are generally found in positions that resist tensile loading. In animals, cells produce fibril-forming collagen molecules that self-assemble into larger structures known as collagen fibrils. Collagen fibrils are the fundamental, continuous, load-bearing elements in connective tissues, but are often further aggregated into larger load-bearing structures, fascicles in tendon, lamellae in cornea and in intervertebral disk. We know that failure to form fibrillar collagen is embryonic lethal, and excessive collagen formation/growth (fibrosis) or uncontrolled enzymatic remodeling (type II collagen: osteoarthritis) is pathological. Collagen is thus critical to vertebrate viability and instrumental in maintaining efficient mechanical structures. However, despite decades of research, our understanding of collagen matrix formation is not complete, and we know still less about the detailed mechanisms that drive collagen remodeling, growth, and pathology. In this perspective, we examine the known role of mechanical force on the formation and development of collagenous structure. We then discuss a mechanochemical mechanism that has the potential to unify our understanding of collagenous tissue assembly dynamics, which preferentially deposits and grows collagen fibrils directly in the path of mechanical force, where the energetics should be dissuasive and where collagen fibrils are most required. We term this mechanism: Mechanochemical force-structure causality. STATEMENT OF SIGNIFICANCE: Our mechanochemical-force structure causality postulate suggests that collagen molecules are components of mechanochemically-sensitive and dynamically-responsive fibrils. Collagen molecules assemble preferentially in the path of applied strain, can be grown in place by mechanical extension, and are retained in the path of force through strain-stabilization. The mechanisms that drive this behavior operate at the level of the molecules themselves and are encoded into the structure of the biomaterial. The concept might change our understanding of structure formation, enhance our ability to treat injuries, and accelerate the development of therapeutics to prevent pathologies such as fibrosis. We suggest that collagen is a mechanochemically responsive dynamic element designed to provide a substantial "material assist" in the construction of adaptive carriers of mechanical signals.
Collapse
|
6
|
Wu C, Cook R, Wu P, Srikumar N, Chee E, Sawyer WS, Wang H, Hazen M, Hotzel I, N'Diaye EN, Ding N, Liu Y, Tran JC, Ye Z. Using a peptide-based mass spectrometry approach to quantitate proteolysis of an intact heterogeneous procollagen substrate by BMP1 for antagonistic antibody screening. Anal Bioanal Chem 2022; 414:6601-6610. [PMID: 35821276 DOI: 10.1007/s00216-022-04220-2] [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: 04/24/2022] [Revised: 06/24/2022] [Accepted: 06/30/2022] [Indexed: 11/01/2022]
Abstract
Proteases are critical proteins involved in cleaving substrates that may impact biological pathways, cellular processes, or disease progression. In the biopharmaceutical industry, modulating the levels of protease activity is an important strategy for mitigating many types of diseases. While a variety of analytical tools exist for characterizing substrate cleavages, in vitro functional screening for antibody inhibitors of protease activity using physiologically relevant intact protein substrates remains challenging. In addition, detecting such large protein substrates with high heterogeneity using high-throughput mass spectrometry screening has rarely been reported in the literature with concerns for assay robustness and sensitivity. In this study, we established a peptide-based in vitro functional screening assay for antibody inhibitors of mouse bone morphogenic protein 1 (mBMP1) metalloprotease using a heterogeneous recombinant 66-kDa mouse Procollagen I alpha 1 chain (mProcollagen) substrate. We compared several analytical tools including capillary gel electrophoresis Western blot (CE-Western blot), as well as both intact protein and peptide-based mass spectrometry (MS) to quantitate the mBMP1 proteolytic activity and its inhibition by antibodies using this heterogeneous mProcollagen substrate. We concluded that the peptide-based mass spectrometry screening assay was the most suitable approach in terms of throughput, sensitivity, and assay robustness. We then optimized our mBMP1 proteolysis reaction after characterizing the enzyme kinetics using the peptide-based MS assay. This assay resulted in Z' values ranging from 0.6 to 0.8 from the screening campaign. Among over 1200 antibodies screened, IC50 characterization was performed on the top candidate hits, which showed partial or complete inhibitory activities against mBMP1.
Collapse
Affiliation(s)
- Cong Wu
- Department of Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA.
| | - Ryan Cook
- Department of Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA
| | - Ping Wu
- Department of Structural Biology, Genentech, South San Francisco, CA, USA
| | | | - Elin Chee
- Department of Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA
| | - William S Sawyer
- Department of Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA
| | - Hua Wang
- Morphic Therapeutic, Waltham, MA, USA
| | - Meredith Hazen
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Isidro Hotzel
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
| | | | - Ning Ding
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | | | - John C Tran
- Department of Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA
| | - Zhengmao Ye
- Department of Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA
| |
Collapse
|
7
|
Ma HY, N'Diaye EN, Caplazi P, Huang Z, Arlantico A, Jeet S, Wong A, Brightbill HD, Li Q, Wong WR, Sandoval W, Tam L, Newman R, Roose-Girma M, Ding N. BMP1 is not required for lung fibrosis in mice. Sci Rep 2022; 12:5466. [PMID: 35361882 PMCID: PMC8971496 DOI: 10.1038/s41598-022-09557-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/24/2022] [Indexed: 01/01/2023] Open
Abstract
Bone morphogenetic protein 1 (BMP1) belongs to the astacin/BMP1/tolloid-like family of zinc metalloproteinases, which play a fundamental role in the development and formation of extracellular matrix (ECM). BMP1 mediates the cleavage of carboxyl terminal (C-term) propeptides from procollagens, a crucial step in fibrillar collagen fiber formation. Blocking BMP1 by small molecule or antibody inhibitors has been linked to anti-fibrotic activity in the preclinical models of skin, kidney and liver fibrosis. Therefore, we reason that BMP1 may be important for the pathogenesis of lung fibrosis and BMP1 could be a potential therapeutic target for progressive fibrotic disease such as idiopathic pulmonary fibrosis (IPF). Here, we observed the increased expression of BMP1 in both human IPF lungs and mouse fibrotic lungs induced by bleomycin. Furthermore, we developed an inducible Bmp1 conditional knockout (cKO) mouse strain. We found that Bmp1 deletion does not protect mice from lung fibrosis triggered by bleomycin. Moreover, we found no significant impact of BMP1 deficiency upon C-term propeptide of type I procollagen (CICP) production in the fibrotic mouse lungs. Based on these results, we propose that BMP1 is not required for lung fibrosis in mice and BMP1 may not be considered a candidate therapeutic target for IPF.
Collapse
Affiliation(s)
- Hsiao-Yen Ma
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Elsa-Noah N'Diaye
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA
| | - Patrick Caplazi
- Department of Pathology, Genentech, South San Francisco, CA, USA
| | - Zhiyu Huang
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Alexander Arlantico
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Surinder Jeet
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Aaron Wong
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Hans D Brightbill
- Department of Translational Immunology, Genentech, South San Francisco, CA, USA
| | - Qingling Li
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, CA, USA
| | - Weng Ruth Wong
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, CA, USA
| | - Wendy Sandoval
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, CA, USA
| | - Lucinda Tam
- Department of Molecular Biology, Genentech, South San Francisco, CA, USA
| | - Robert Newman
- Department of Molecular Biology, Genentech, South San Francisco, CA, USA
| | - Merone Roose-Girma
- Department of Molecular Biology, Genentech, South San Francisco, CA, USA
| | - Ning Ding
- Department of Discovery Immunology, Genentech, South San Francisco, CA, USA.
| |
Collapse
|
8
|
Siadat SM, Zamboulis DE, Thorpe CT, Ruberti JW, Connizzo BK. Tendon Extracellular Matrix Assembly, Maintenance and Dysregulation Throughout Life. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1348:45-103. [PMID: 34807415 DOI: 10.1007/978-3-030-80614-9_3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In his Lissner Award medal lecture in 2000, Stephen Cowin asked the question: "How is a tissue built?" It is not a new question, but it remains as relevant today as it did when it was asked 20 years ago. In fact, research on the organization and development of tissue structure has been a primary focus of tendon and ligament research for over two centuries. The tendon extracellular matrix (ECM) is critical to overall tissue function; it gives the tissue its unique mechanical properties, exhibiting complex non-linear responses, viscoelasticity and flow mechanisms, excellent energy storage and fatigue resistance. This matrix also creates a unique microenvironment for resident cells, allowing cells to maintain their phenotype and translate mechanical and chemical signals into biological responses. Importantly, this architecture is constantly remodeled by local cell populations in response to changing biochemical (systemic and local disease or injury) and mechanical (exercise, disuse, and overuse) stimuli. Here, we review the current understanding of matrix remodeling throughout life, focusing on formation and assembly during the postnatal period, maintenance and homeostasis during adulthood, and changes to homeostasis in natural aging. We also discuss advances in model systems and novel tools for studying collagen and non-collagenous matrix remodeling throughout life, and finally conclude by identifying key questions that have yet to be answered.
Collapse
Affiliation(s)
| | - Danae E Zamboulis
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Chavaunne T Thorpe
- Comparative Biomedical Sciences, The Royal Veterinary College, University of London, London, UK
| | - Jeffrey W Ruberti
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Brianne K Connizzo
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| |
Collapse
|