1
|
Li M, Deng T, Chen Q, Jiang S, Li H, Li J, You S, Xie HQ, Shen B. A versatile platform based on matrix metalloproteinase-sensitive peptides for novel diagnostic and therapeutic strategies in arthritis. Bioact Mater 2025; 47:100-120. [PMID: 39897588 PMCID: PMC11787566 DOI: 10.1016/j.bioactmat.2025.01.011] [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: 11/25/2024] [Revised: 01/11/2025] [Accepted: 01/12/2025] [Indexed: 02/04/2025] Open
Abstract
Matrix metalloproteinases (MMPs), coupled with other proteinases and glycanases, can degrade proteoglycans, collagens, and other extracellular matrix (ECM) components in inflammatory and non-inflammatory arthritis, making them important pathogenic molecules and ideal disease indicators and pharmaceutical intervention triggers. For MMP responsiveness, MMP-sensitive peptides (MSPs) are among the most easily synthesized and cost-effective substrates, with free terminal amine and/or carboxyl groups extensively employed in multiple designs. We hereby provide a comprehensive review over the mechanisms and advances in MSP applications for the management of arthritis. These applications include early and precise diagnosis of MMP activity via fluorescence probe technologies; acting as nanodrug carriers to enable on-demand drug release triggered by pathological microenvironments; and facilitating cartilage engineering through MMP-mediated degradation, which promotes cell migration, matrix synthesis, and tissue integration. Specifically, the ultra-sensitive MSP diagnostic probes could significantly advance the early diagnosis and detection of osteoarthritis (OA), while MSP-based drug carriers for rheumatoid arthritis (RA) can intelligently release anti-inflammatory drugs effectively during flare-ups, or even before symptoms manifest. The continuous progress in MSP development may acceleratedly lead to novel management regimens for arthropathy in the future.
Collapse
Affiliation(s)
- Mingyang Li
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tao Deng
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Quan Chen
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Shenghu Jiang
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hang Li
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiayi Li
- Department of Nephrology, The People's Hospital of Yubei District of Chongqing, Chongqing, China
| | - Shenglan You
- Animal Imaging Core Facilities, West China Hospital, Sichuan University, China
| | - Hui-qi Xie
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Stem Cell and Tissue Engineering Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bin Shen
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
2
|
Edwards SD, Tian Z, Kim YJ, Jeong KJ. Sepia Melanin for the Local Deactivation of Matrix Metalloproteinases. ACS Macro Lett 2025:538-543. [PMID: 40233351 DOI: 10.1021/acsmacrolett.4c00792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent enzymes that degrade the extracellular matrix and play essential functions in wound healing and tissue remodeling. However, the long-term overexpression of MMPs is associated with a chronic wound environment. Treatments that locally deactivate MMPs have been proposed to improve the outcomes of nonhealing chronic wounds. In this Letter, sepia melanin, a natural biopigment, is explored for the deactivation of MMPs, through chelation of zinc ions. Melanins have chemical functional groups to bind to various metal ions, which we demonstrate can be exploited for the deactivation of MMPs. Melanin's selectivity for zinc ions, and ability to deactivate MMPs that are associated with chronic wounds (MMP-1, MMP-2, and MMP-9) are examined, and its potential application in wound healing is demonstrated by functional in vitro tissue assays, mimicking the nonhealing wound environment. This is the first demonstration of the use of melanin for MMP deactivation.
Collapse
Affiliation(s)
- Seth D Edwards
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, New Hampshire 03824, United States
| | - Zhen Tian
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, New Hampshire 03824, United States
| | - Young Jo Kim
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, New Hampshire 03824, United States
| | - Kyung Jae Jeong
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, New Hampshire 03824, United States
| |
Collapse
|
3
|
Diaz JA, Gianesini S, Khalil RA. Glycocalyx disruption, endothelial dysfunction and vascular remodeling as underlying mechanisms and treatment targets of chronic venous disease. INT ANGIOL 2024; 43:563-590. [PMID: 39873224 PMCID: PMC11839207 DOI: 10.23736/s0392-9590.24.05339-2] [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] [Indexed: 01/30/2025]
Abstract
The glycocalyx is an essential structural and functional component of endothelial cells. Extensive hemodynamic changes cause endothelial glycocalyx disruption and vascular dysfunction, leading to multiple arterial and venous disorders. Chronic venous disease (CVD) is a common disorder of the lower extremities with major health and socio-economic implications, but complex pathophysiology. Genetic aberrations accentuated by environmental factors, behavioral tendencies, and hormonal disturbances promote venous reflux, valve incompetence, and venous blood stasis. Increased venous hydrostatic pressure and changes in shear-stress cause glycocalyx injury, endothelial dysfunction, secretion of adhesion molecules, leukocyte recruitment/activation, and release of cytokines, chemokines, and hypoxia-inducible factor, causing smooth muscle cell switch from contractile to synthetic proliferative phenotype, imbalance in matrix metalloproteinases (MMPs), degradation of collagen and elastin, and venous tissue remodeling, leading to venous dilation and varicose veins. In the advanced stages of CVD, leukocyte infiltration of the vein wall causes progressive inflammation, fibrosis, disruption of junctional proteins, accumulation of tissue metabolites and reactive oxygen and nitrogen species, and iron deposition, leading to skin changes and venous leg ulcer (VLU). CVD management includes compression stockings, venotonics, and surgical intervention. In addition to its antithrombotic and fibrinolytic properties, literature suggests sulodexide benefits in reducing inflammation, promoting VLU healing, improving endothelial function, exhibiting venotonic properties, and inhibiting MMP-9. Understanding the role of glycocalyx, endothelial dysfunction, and vascular remodeling should help delineate the underlying mechanisms and develop improved biomarkers and targeted therapy for CVD and VLU.
Collapse
Affiliation(s)
- Jose A. Diaz
- Division of Surgical Research, Light Surgical Research and Training Laboratory, Surgical Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sergio Gianesini
- Vascular Diseases Center, Translational Surgery Unit, University of Ferrara, Ferrara, Italy, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Raouf A. Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
4
|
Roy HS, Murugesan P, Kulkarni C, Arora M, Nagar GK, Guha R, Chattopadhyay N, Ghosh D. On-demand release of a selective MMP-13 blocker from an enzyme-responsive injectable hydrogel protects cartilage from degenerative progression in osteoarthritis. J Mater Chem B 2024; 12:5325-5338. [PMID: 38669084 DOI: 10.1039/d3tb02871b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
In osteoarthritis (OA), the degradation of cartilage is primarily driven by matrix metalloprotease-13 (MMP-13). Hence, the inhibition of MMP-13 has emerged as an attractive target for OA treatment. Among the various approaches that are being explored for MMP-13 regulation, blocking of the enzyme with specific binding molecules appears to be a more promising strategy for preventing cartilage degeneration. To enhance effectiveness and ensure patient compliance, it is preferable for the binding molecule to exhibit sustained activity when administered directly into the joint. Herein, we present an enzyme-responsive hydrogel that was designed to exhibit on-demand, the sustained release of BI-4394, a potent and highly selective MMP-13 blocker. The stable and compatible hydrogel was prepared using triglycerol monostearate. The efficacy of the hydrogel to prevent cartilage damage was assessed in a rat model of OA induced by anterior cruciate ligament transection (ACLT). The results revealed that in comparison to the rats administrated weekly with intra-articular BI-4394, the hydrogel implanted rats had reduced levels of inflammation and bone erosion. In comparison to untreated control, the cartilage in animals administered with BI-4394/hydrogel exhibited significant levels of collagen-2 and aggrecan along with reduced MMP-13. Overall, this study confirmed the potential of BI-4394 delivery using an enzyme-responsive hydrogel as a promising treatment option to treat the early stages of OA by preventing further cartilage degradation.
Collapse
Affiliation(s)
- Himadri Shekhar Roy
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Preethi Murugesan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Chirag Kulkarni
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Malika Arora
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Geet Kumar Nagar
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Rajdeep Guha
- Division of Laboratory Animal Facility, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Deepa Ghosh
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| |
Collapse
|
5
|
Moser MS, Hallem EA. Astacin metalloproteases in human-parasitic nematodes. ADVANCES IN PARASITOLOGY 2024; 126:177-204. [PMID: 39448190 DOI: 10.1016/bs.apar.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Parasitic nematodes infect over 2 billion individuals worldwide, primarily in low-resource areas, and are responsible for several chronic and potentially deadly diseases. Throughout their life cycle, these parasites are thought to use astacin metalloproteases, a subfamily of zinc-containing metalloendopeptidases, for processes such as skin penetration, molting, and tissue migration. Here, we review the known functions of astacins in human-infective, soil-transmitted parasitic nematodes - including the hookworms Necator americanus and Ancylostoma duodenale, the threadworm Strongyloides stercoralis, the giant roundworm Ascaris lumbricoides, and the whipworm Trichuris trichiura - as well as the human-infective, vector-borne filarial nematodes Wuchereria bancrofti, Onchocerca volvulus, and Brugia malayi. We also review astacin function in parasitic nematodes that infect other mammalian hosts and discuss the potential of astacins as anthelmintic drug targets. Finally, we highlight the molecular and genetic tools that are now available for further exploration of astacin function and discuss how a better understanding of astacin function in human-parasitic nematodes could lead to new avenues for nematode control and drug therapies.
Collapse
Affiliation(s)
- Matthew S Moser
- Molecular Biology Interdepartmental PhD Program; Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Elissa A Hallem
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States.
| |
Collapse
|
6
|
Grillet B, Pereira RVS, Van Damme J, Abu El-Asrar A, Proost P, Opdenakker G. Matrix metalloproteinases in arthritis: towards precision medicine. Nat Rev Rheumatol 2023; 19:363-377. [PMID: 37161083 DOI: 10.1038/s41584-023-00966-w] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2023] [Indexed: 05/11/2023]
Abstract
Proteolysis of structural molecules of the extracellular matrix (ECM) is an irreversible post-translational modification in all arthropathies. Common joint disorders, including osteoarthritis and rheumatoid arthritis, have been associated with increased levels of matrix remodelling enzymes, including matrix metalloproteinases (MMPs). MMPs, in concert with other host proteinases and glycanases, destroy proteoglycans, collagens and other ECM molecules. MMPs may also control joint remodelling indirectly by signalling through cell-surface receptors or by proteolysis of cytokines and receptor molecules. After synthesis as pro-forms, MMPs can be activated by various types of post-translational modifications, including proteolysis. Once activated, MMPs are controlled by general and specific tissue inhibitors of metalloproteinases (TIMPs). In rheumatoid arthritis, proteolysis of the ECM results in so-called remnant epitopes that enhance and perpetuate autoimmune processes in susceptible hosts. In osteoarthritis, the considerable production of MMP-13 by chondrocytes, often concurrent with mechanical overload, is a key event. Hence, information about the regulation, timing, localization and activities of MMPs in specific disease phases and arthritic entities will help to develop better diagnostics. Insights into beneficial and detrimental effects of MMPs on joint tissue inflammation are also necessary to plan and execute (pre)clinical studies for better therapy and precision medicine with MMP inhibitors. With the advances in proteomics and single-cell transcriptomics, two critical points need attention: neglected neutrophil MMP biology, and the analysis of net proteolytic activities as the result of balances between MMPs and their inhibitors.
Collapse
Affiliation(s)
- Bernard Grillet
- Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Rafaela Vaz Sousa Pereira
- Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Jo Van Damme
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Ahmed Abu El-Asrar
- Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Ophthalmology, King Saud University, Riyadh, Saudi Arabia
| | - Paul Proost
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium.
- Department of Ophthalmology, King Saud University, Riyadh, Saudi Arabia.
- University Hospitals Gasthuisberg, UZ Leuven, KU Leuven, Leuven, Belgium.
| |
Collapse
|
7
|
Fan A, Wu G, Wang J, Lu L, Wang J, Wei H, Sun Y, Xu Y, Mo C, Zhang X, Pang Z, Pan Z, Wang Y, Lu L, Fu G, Ma M, Zhu Q, Cao D, Qin J, Yin F, Yue R. Inhibition of fibroblast activation protein ameliorates cartilage matrix degradation and osteoarthritis progression. Bone Res 2023; 11:3. [PMID: 36588124 PMCID: PMC9806108 DOI: 10.1038/s41413-022-00243-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 10/14/2022] [Accepted: 11/11/2022] [Indexed: 01/03/2023] Open
Abstract
Fibroblast activation protein (Fap) is a serine protease that degrades denatured type I collagen, α2-antiplasmin and FGF21. Fap is highly expressed in bone marrow stromal cells and functions as an osteogenic suppressor and can be inhibited by the bone growth factor Osteolectin (Oln). Fap is also expressed in synovial fibroblasts and positively correlated with the severity of rheumatoid arthritis (RA). However, whether Fap plays a critical role in osteoarthritis (OA) remains poorly understood. Here, we found that Fap is significantly elevated in osteoarthritic synovium, while the genetic deletion or pharmacological inhibition of Fap significantly ameliorated posttraumatic OA in mice. Mechanistically, we found that Fap degrades denatured type II collagen (Col II) and Mmp13-cleaved native Col II. Intra-articular injection of rFap significantly accelerated Col II degradation and OA progression. In contrast, Oln is expressed in the superficial layer of articular cartilage and is significantly downregulated in OA. Genetic deletion of Oln significantly exacerbated OA progression, which was partially rescued by Fap deletion or inhibition. Intra-articular injection of rOln significantly ameliorated OA progression. Taken together, these findings identify Fap as a critical pathogenic factor in OA that could be targeted by both synthetic and endogenous inhibitors to ameliorate articular cartilage degradation.
Collapse
Affiliation(s)
- Aoyuan Fan
- grid.24516.340000000123704535Department of Joint Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092 China
| | - Genbin Wu
- grid.16821.3c0000 0004 0368 8293Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240 China
| | - Jianfang Wang
- grid.24516.340000000123704535Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092 China
| | - Laiya Lu
- grid.24516.340000000123704535Department of Joint Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092 China
| | - Jingyi Wang
- grid.24516.340000000123704535Department of Joint Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092 China
| | - Hanjing Wei
- grid.24516.340000000123704535Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092 China
| | - Yuxi Sun
- grid.24516.340000000123704535Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Yanhua Xu
- grid.24516.340000000123704535Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092 China ,grid.24516.340000000123704535Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Chunyang Mo
- grid.24516.340000000123704535Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092 China
| | - Xiaoying Zhang
- grid.24516.340000000123704535Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092 China
| | - Zhiying Pang
- grid.24516.340000000123704535Department of Joint Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092 China
| | - Zhangyi Pan
- grid.24516.340000000123704535Department of Joint Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092 China
| | - Yiming Wang
- grid.24516.340000000123704535Department of Joint Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092 China
| | - Liangyu Lu
- grid.24516.340000000123704535Department of Joint Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092 China
| | - Guojian Fu
- grid.16821.3c0000 0004 0368 8293Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240 China
| | - Mengqiu Ma
- grid.24516.340000000123704535Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Qiaoling Zhu
- grid.24516.340000000123704535Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092 China
| | - Dandan Cao
- grid.24516.340000000123704535Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092 China
| | - Jiachen Qin
- grid.24516.340000000123704535Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092 China
| | - Feng Yin
- grid.24516.340000000123704535Department of Joint Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092 China ,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120 China ,grid.452344.0Shanghai Clinical Research Center for Aging and Medicine, Shanghai, 200040 China
| | - Rui Yue
- grid.24516.340000000123704535Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092 China ,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120 China
| |
Collapse
|
8
|
Baidya SK, Banerjee S, Adhikari N, Jha T. Selective Inhibitors of Medium-Size S1' Pocket Matrix Metalloproteinases: A Stepping Stone of Future Drug Discovery. J Med Chem 2022; 65:10709-10754. [PMID: 35969157 DOI: 10.1021/acs.jmedchem.1c01855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Among various matrix metalloproteinases (MMPs), MMPs having medium-size S1' pockets are established as promising biomolecular targets for executing crucial roles in cancer, cardiovascular diseases, and neurodegenerative diseases. However, no such MMP inhibitors (MMPIs) are available to date as drug candidates despite a lot of continuous research work for more than three decades. Due to a high degree of structural resemblance among these MMPs, designing selective MMPIs is quite challenging. However, the variability and uniqueness of the S1' pockets of these MMPs make them promising targets for designing selective MMPIs. In this perspective, the overall structural aspects of medium-size S1' pocket MMPs including the unique binding patterns of enzyme-inhibitor interactions have been discussed in detail to acquire knowledge regarding selective inhibitor designing. This overall knowledge will surely be a curtain raiser for the designing of selective MMPIs as drug candidates in the future.
Collapse
Affiliation(s)
- Sandip Kumar Baidya
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Suvankar Banerjee
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Nilanjan Adhikari
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| |
Collapse
|
9
|
Halland N, Schmidt F, Weiss T, Li Z, Czech J, Saas J, Ding-Pfennigdorff D, Dreyer MK, Strübing C, Nazare M. Rational Design of Highly Potent, Selective, and Bioavailable SGK1 Protein Kinase Inhibitors for the Treatment of Osteoarthritis. J Med Chem 2021; 65:1567-1584. [PMID: 34931844 DOI: 10.1021/acs.jmedchem.1c01601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The serine/threonine kinase SGK1 is an activator of the β-catenin pathway and a powerful stimulator of cartilage degradation that is found to be upregulated under genomic control in diseased osteoarthritic cartilage. Today, no oral disease-modifying treatments are available and chronic treatment in this indication sets high requirements for the drug selectivity, pharmacokinetic, and safety profile. We describe the identification of a highly selective druglike 1H-pyrazolo[3,4-d]pyrimidine SGK1 inhibitor 17a that matches both safety and pharmacokinetic requirements for oral dosing. Rational compound design was facilitated by a novel hSGK1 co-crystal structure, and multiple ligand-based computer models were applied to guide the chemical optimization of the compound ADMET and selectivity profiles. Compounds were selected for subchronic proof of mechanism studies in the mouse femoral head cartilage explant model, and compound 17a emerged as a druglike SGK1 inhibitor, with a highly optimized profile suitable for oral dosing as a novel, potentially disease-modifying agent for osteoarthritis.
Collapse
Affiliation(s)
- Nis Halland
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Friedemann Schmidt
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Tilo Weiss
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Ziyu Li
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Jörg Czech
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Joachim Saas
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | | | - Matthias K Dreyer
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Carsten Strübing
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Marc Nazare
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125 Berlin-Buch, Germany
| |
Collapse
|
10
|
Bendele AM, Neelagiri M, Neelagiri V, Sucholeiki I. Development of a selective matrix metalloproteinase 13 (MMP-13) inhibitor for the treatment of Osteoarthritis. Eur J Med Chem 2021; 224:113666. [PMID: 34245949 PMCID: PMC8511113 DOI: 10.1016/j.ejmech.2021.113666] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 04/07/2021] [Accepted: 06/13/2021] [Indexed: 01/15/2023]
Abstract
Osteoarthritis (OA) is a chronic disorder that causes damage to the cartilage and surrounding tissues and is characterized by pain, stiffness, and loss of function. Current treatments for OA primarily involve providing only relief of symptoms but does not affect the overall trajectory of the disease. A major goal for treating OA has been to slow down or reverse disease progression. Matrix metalloproteinase-13 (MMP-13) is expressed by chondrocytes and synovial cells in human OA and is thought to play a critical role in cartilage destruction. Herein we report a new, allosteric MMP-13 inhibitor, AQU-019, that has been optimized for potency, metabolic stability, and oral bioavailability through a combination of structure activity relationship (SAR) and deuterium substitution as a potential disease modifying OA drug (DMOAD). The inhibitor was demonstrated to be chondroprotective when injected intraarticular (IA) in the monoiodoacetic acid (MIA) rat model of OA.
Collapse
Affiliation(s)
- Alison M Bendele
- BolderBioPath, Inc., 5541 Central Avenue, Suite 160, Boulder, CO, 80301, USA
| | - Madhavi Neelagiri
- Aquilus Pharmaceuticals Inc., 400 West Cummings Park, Woburn, MA, 01801, USA
| | - Venugopal Neelagiri
- Aquilus Pharmaceuticals Inc., 400 West Cummings Park, Woburn, MA, 01801, USA
| | - Irving Sucholeiki
- Aquilus Pharmaceuticals Inc., 400 West Cummings Park, Woburn, MA, 01801, USA.
| |
Collapse
|
11
|
Kućmierz J, Frąk W, Młynarska E, Franczyk B, Rysz J. Molecular Interactions of Arterial Hypertension in Its Target Organs. Int J Mol Sci 2021; 22:ijms22189669. [PMID: 34575833 PMCID: PMC8471598 DOI: 10.3390/ijms22189669] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/30/2021] [Accepted: 09/03/2021] [Indexed: 02/08/2023] Open
Abstract
Arterial hypertension (AH) is a major risk factor for the development of cardiovascular diseases. It is estimated that the disease affects between 10% and 20% of the adult population and is responsible for 5.8% of all deaths worldwide. Several pathophysiologic factors are crucial in AH, including inappropriate activation of the renin-angiotensin-aldosterone system, oxidative stress and inflammation. The heart, kidney, brain, retina and arterial blood vessels are prime targets of hypertensive damage. Uncontrolled and untreated AH accelerates the damage to these organs and could cause their failure. Damage to these organs could also manifest as coronary heart disease, cognitive impairment, retinopathy or optic neuropathy. For better understanding, it is important to analyze molecular factors which take part in pathogenesis of AH and hypertension-related target organ damage. In our paper, we would like to focus on molecular interactions of AH in the heart, blood vessels, brain and kidneys. We focus on matrix metalloproteinases, the role of immune system, the renin-angiotensin-aldosterone system and oxidative stress in hypertensive induced organ damage.
Collapse
|
12
|
Identification of Broad-Spectrum MMP Inhibitors by Virtual Screening. Molecules 2021; 26:molecules26154553. [PMID: 34361703 PMCID: PMC8347235 DOI: 10.3390/molecules26154553] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 12/20/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are the family of proteases that are mainly responsible for degrading extracellular matrix (ECM) components. In the skin, the overexpression of MMPs as a result of ultraviolet radiation triggers an imbalance in the ECM turnover in a process called photoaging, which ultimately results in skin wrinkling and premature skin ageing. Therefore, the inhibition of different enzymes of the MMP family at a topical level could have positive implications for photoaging. Considering that the MMP catalytic region is mostly conserved across different enzymes of the MMP family, in this study we aimed to design a virtual screening (VS) workflow to identify broad-spectrum MMP inhibitors that can be used to delay the development of photoaging. Our in silico approach was validated in vitro with 20 VS hits from the Specs library that were not only structurally different from one another but also from known MMP inhibitors. In this bioactivity assay, 18 of the 20 compounds inhibit at least one of the assayed MMPs at 100 μM (with 5 of them showing around 50% inhibition in all the tested MMPs at this concentration). Finally, this VS was used to identify natural products that have the potential to act as broad-spectrum MMP inhibitors and be used as a treatment for photoaging.
Collapse
|
13
|
Raffetto JD, Khalil RA. Mechanisms of Lower Extremity Vein Dysfunction in Chronic Venous Disease and Implications in Management of Varicose Veins. VESSEL PLUS 2021; 5. [PMID: 34250453 DOI: 10.20517/2574-1209.2021.16] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Chronic venous disease (CVD) is a common venous disorder of the lower extremities. CVD can be manifested as varicose veins (VVs), with dilated and tortuous veins, dysfunctional valves and venous reflux. If not adequately treated, VVs could progress to chronic venous insufficiency (CVI) and lead to venous leg ulcer (VLU). Predisposing familial and genetic factors have been implicated in CVD. Additional environmental, behavioral and dietary factors including sedentary lifestyle and obesity may also contribute to CVD. Alterations in the mRNA expression, protein levels and proteolytic activity of matrix metalloproteinases (MMPs) have been detected in VVs and VLU. MMP expression/activity can be modulated by venous hydrostatic pressure, hypoxia, tissue metabolites, and inflammation. MMPs in turn increase proteolysis of different protein substrates in the extracellular matrix particularly collagen and elastin, leading to weakening of the vein wall. MMPs could also promote venous dilation by increasing the release of endothelium-derived vasodilators and activating potassium channels, leading to smooth muscle hyperpolarization and relaxation. Depending on VVs severity, management usually includes compression stockings, sclerotherapy and surgical removal. Venotonics have also been promoted to decrease the progression of VVs. Sulodexide has also shown benefits in VLU and CVI, and recent data suggest that it could improve venous smooth muscle contraction. Other lines of treatment including induction of endogenous tissue inhibitors of metalloproteinases (TIMPs) and administration of exogenous synthetic inhibitors of MMPs are being explored, and could provide alternative strategies in the treatment of CVD.
Collapse
Affiliation(s)
- Joseph D Raffetto
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA
| |
Collapse
|
14
|
Mlost J, Kostrzewa M, Borczyk M, Bryk M, Chwastek J, Korostyński M, Starowicz K. CB2 agonism controls pain and subchondral bone degeneration induced by mono-iodoacetate: Implications GPCR functional bias and tolerance development. Biomed Pharmacother 2021; 136:111283. [PMID: 33482616 DOI: 10.1016/j.biopha.2021.111283] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE The endocannabinoid system became a promising target for osteoarthritis (OA) treatment. Functional selectivity of cannabinoids may increase their beneficial properties while reducing side effects. The aim of the present study was to evaluate the analgesic potential of two functionally biased CB2 agonists in different treatment regimens to propose the best pharmacological approach for OA management. EXPERIMENTAL APPROACH Two functionally selective CB2 agonists were administered i.p. - JWH133 (cAMP biased) and GW833972A (β-arrestin biased), in a chemically induced model of OA in rats. The drugs were tested in acute and chronic treatment regimens. Analgesic effects were assessed by pressure application measurement and kinetic weight bearing. X-ray microtomography was used for the morphometric analysis of the femur's subchondral bone tissue. Underlying biochemical changes were analysed via RT-qPCR. KEY RESULTS Dose-response studies established the effective dose for both JWH133 and GW833972A. In chronic treatment paradigms, JWH133 was able to elicit analgesia throughout the course of the experiment, whereas GW833972A lost its efficacy after 2 days of treatment. Later studies revealed improvement in subchondral bone architecture and decrement of matrix metalloproteinases and proinflammatory factors expression following JWH133 chronic treatment. CONCLUSION AND IMPLICATIONS Data presents analgesic and disease-modifying potential of CB2 agonists in OA treatment. Moreover, the study revealed more pronounced tolerance development for analgesic effects of the β-arrestin biased CB2 agonist GW833972A. These results provide a better understanding of the molecular underpinnings of the anti-nociceptive potential of CB2 agonists and may improve drug development processes for any cannabinoid-based chronic pain therapy.
Collapse
Affiliation(s)
- Jakub Mlost
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Cracow, Poland
| | - Magdalena Kostrzewa
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Cracow, Poland
| | - Małgorzata Borczyk
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Cracow, Poland
| | - Marta Bryk
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Cracow, Poland
| | - Jakub Chwastek
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Cracow, Poland
| | - Michał Korostyński
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Cracow, Poland
| | - Katarzyna Starowicz
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Cracow, Poland.
| |
Collapse
|
15
|
Hu Q, Ecker M. Overview of MMP-13 as a Promising Target for the Treatment of Osteoarthritis. Int J Mol Sci 2021; 22:ijms22041742. [PMID: 33572320 PMCID: PMC7916132 DOI: 10.3390/ijms22041742] [Citation(s) in RCA: 212] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 01/02/2023] Open
Abstract
Osteoarthritis (OA) is a common degenerative disease characterized by the destruction of articular cartilage and chronic inflammation of surrounding tissues. Matrix metalloproteinase-13 (MMP-13) is the primary MMP involved in cartilage degradation through its particular ability to cleave type II collagen. Hence, it is an attractive target for the treatment of OA. However, the detailed molecular mechanisms of OA initiation and progression remain elusive, and, currently, there are no interventions available to restore degraded cartilage. This review fully illustrates the involvement of MMP-13 in the initiation and progression of OA through the regulation of MMP-13 activity at the molecular and epigenetic levels, as well as the strategies that have been employed against MMP-13. The aim of this review is to identify MMP-13 as an attractive target for inhibitor development in the treatment of OA.
Collapse
|
16
|
Pijet B, Konopka A, Rejmak E, Stefaniuk M, Khomiak D, Bulska E, Pikul S, Kaczmarek L. The matrix metalloproteinase inhibitor marimastat inhibits seizures in a model of kainic acid-induced status epilepticus. Sci Rep 2020; 10:21314. [PMID: 33277582 PMCID: PMC7718901 DOI: 10.1038/s41598-020-78341-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
An intra-hippocampus injection of kainic acid serves as a model of status epilepticus and the subsequent development of temporal lobe epilepsy. Matrix metalloproteinase-9 (MMP-9) is an enzyme that controls remodeling of the extracellular milieu under physiological and pathological conditions. In response to brain insult, MMP-9 contributes to pathological synaptic plasticity that may play a role in the progression of an epileptic condition. Marimastat is a metalloproteinase inhibitor that was tested in clinical trials of cancer. The present study assessed whether marimastat can impair the development of epilepsy. The inhibitory efficacy of marimastat was initially tested in neuronal cultures in vitro. As a marker substrate, we used nectin-3. Next, we investigated the blood–brain barrier penetration of marimastat using mass spectrometry and evaluated the therapeutic potential of marimastat against seizure outcomes. We found that marimastat inhibited the cleavage of nectin-3 in hippocampal neuronal cell cultures. Marimastat penetrated the blood–brain barrier and exerted an inhibitory effect on metalloproteinase activity in the brain. Finally, marimastat decreased some seizure parameters, such as seizure score and number, but did not directly affect status epilepticus. The long-term effects of marimastat were evident up to 6 weeks after kainic acid administration, in which marimastat still inhibited seizure duration.
Collapse
Affiliation(s)
- Barbara Pijet
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland.
| | - Anna Konopka
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-093, Warsaw, Poland
| | - Emilia Rejmak
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Marzena Stefaniuk
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Danylo Khomiak
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Ewa Bulska
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-093, Warsaw, Poland
| | - Stanisław Pikul
- Pikralida Sp. z o.o., Bukowska 70/b424, 60-812, Poznań, Poland
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| |
Collapse
|
17
|
Cuffaro D, Nuti E, D’Andrea F, Rossello A. Developments in Carbohydrate-Based Metzincin Inhibitors. Pharmaceuticals (Basel) 2020; 13:ph13110376. [PMID: 33182755 PMCID: PMC7696829 DOI: 10.3390/ph13110376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/03/2020] [Accepted: 11/06/2020] [Indexed: 01/03/2023] Open
Abstract
Matrix metalloproteinases (MMPs) and A disintegrin and Metalloproteinase (ADAMs) are zinc-dependent endopeptidases belonging to the metzincin superfamily. Upregulation of metzincin activity is a major feature in many serious pathologies such as cancer, inflammations, and infections. In the last decades, many classes of small molecules have been developed directed to inhibit these enzymes. The principal shortcomings that have hindered clinical development of metzincin inhibitors are low selectivity for the target enzyme, poor water solubility, and long-term toxicity. Over the last 15 years, a novel approach to improve solubility and bioavailability of metzincin inhibitors has been the synthesis of carbohydrate-based compounds. This strategy consists of linking a hydrophilic sugar moiety to an aromatic lipophilic scaffold. This review aims to describe the development of sugar-based and azasugar-based derivatives as metzincin inhibitors and their activity in several pathological models.
Collapse
|
18
|
Roy HS, Dubey G, Sharma VK, Bharatam PV, Ghosh D. Molecular docking and molecular dynamics to identify collagenase inhibitors as lead compounds to address osteoarthritis. J Biomol Struct Dyn 2020; 40:2339-2351. [PMID: 33103592 DOI: 10.1080/07391102.2020.1838326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Osteoarthritis (OA) is a degenerative disease which affects a large number of individuals. Collagenases, which belong to a class of metalloproteases (MMPs), are responsible for the degradation of cartilage manifested in OA. Inhibition of the catalytic domains of these MMPs is one of the important therapeutic strategies proposed for the prevention of OA. The main objective of this work is to evaluate the binding of curcumin and its metabolites with the active sites of collagenases in comparison to standard inhibitors on the basis of our hypothesis that curcumin/metabolites could exhibit an inhibitory effect on MMPs. Here, we report the molecular docking analysis of curcumin and its metabolites with collagenases (MMP-1, MMP-8, MMP-13). Among the molecules tested, curcumin monoglucuronide (CMG) demonstrated the best binding affinity with MMP-13, which is specifically implicated in OA. The CMG-MMP-complexes were further subjected to molecular dynamic simulations to explore the stability of the complexes and to estimate the free binding energies. The results indicated that CMG preferentially bind to MMP-13 in comparison to that of MMP-1 and MMP-8 with binding free energies (ΔGbind) of (-60.55), (-27.02) and (-46.91) kcal/mol, respectively. This is the first study which suggests that curcumin monoglucuronide can be considered as an effective lead compound to prevent the progression of OA.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Himadri Shekhar Roy
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Mohali, Punjab, India
| | - Gurudutt Dubey
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Vishnu Kumar Sharma
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Prasad V Bharatam
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Deepa Ghosh
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Mohali, Punjab, India
| |
Collapse
|
19
|
Fischer T, Riedl R. Challenges with matrix metalloproteinase inhibition and future drug discovery avenues. Expert Opin Drug Discov 2020; 16:75-88. [PMID: 32921161 DOI: 10.1080/17460441.2020.1819235] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Matrix metalloproteinases have been in the scope of pharmaceutical drug discovery for decades as promising targets for drug development. Until present, no modulator of the enzyme class survived clinical trials, all failing for various reasons. Nevertheless, the target family did not lose its attractiveness and there is ever more evidence that MMP modulators are likely to overcome the hurdles and result in successful clinical therapies. AREAS COVERED This review provides an overview of past efforts that were taken in the development of MMP inhibitors and insight into promising strategies that might enable drug discovery in the field in the future. Small molecule inhibitors as well as biomolecules are reviewed. EXPERT OPINION Despite the lack of successful clinical trials in the past, there is ongoing research in the field of MMP modulation, proving the target class has not lost its appeal to pharmaceutical research. With ever-growing insights from different scientific fields that shed light on previously unknown correlations, it is now time to use synergies deriving from biological knowledge, chemical structure generation, and clinical application to reach the ultimate goal of bringing MMP derived drugs on a broad front for the benefit of patients into therapeutic use.
Collapse
Affiliation(s)
- Thomas Fischer
- Center of Organic and Medicinal Chemistry, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences ZHAW , 8820 Wädenswil, Switzerland
| | - Rainer Riedl
- Center of Organic and Medicinal Chemistry, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences ZHAW , 8820 Wädenswil, Switzerland
| |
Collapse
|
20
|
Nakamura H, Vo P, Kanakis I, Liu K, Bou-Gharios G. Aggrecanase-selective tissue inhibitor of metalloproteinase-3 (TIMP3) protects articular cartilage in a surgical mouse model of osteoarthritis. Sci Rep 2020; 10:9288. [PMID: 32518385 PMCID: PMC7283274 DOI: 10.1038/s41598-020-66233-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/18/2020] [Indexed: 01/03/2023] Open
Abstract
A key feature of osteoarthritis is the gradual loss of articular cartilage and bone deformation, resulting in the impairment of joint function. The primary cause of cartilage destruction is considered to be the presence of elevated proteases, such as matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTSs). However, clinically tested global MMP inhibitors have low efficacy that may be due to their lack of selectivity. We previously demonstrated in vitro that a variant of tissue inhibitor of metalloproteinase-3 ([-1A]TIMP3) inhibits ADAMTSs but not MMPs. In this study, we tested whether the selectivity of [-1A]TIMP3 is beneficial compared with that of the wild-type TIMP3 in preventing or delaying the onset of the degenerative effects in a mouse model of osteoarthritis. We generated transgenic mice that overexpressed TIMP3 or [-1A]TIMP3 driven by a chondrocyte-specific type II collagen promoter. TIMP3 transgenic mice showed compromised bone integrity as opposed to [-1A]TIMP3 mice. After surgically induced joint instability, TIMP3 overexpression proved to be less protective in cartilage destruction than [-1A]TIMP3 at late stages of OA. The selective inhibition of ADAMTSs provides the possibility of modifying TIMP3 to specifically target a class of cartilage-degrading proteinases and to minimize adverse effects on bone and possibly other tissues.
Collapse
Affiliation(s)
- Hiroyuki Nakamura
- Department of Oral and Maxillofacial Surgery, Kanazawa University Graduate School of Medical Science Kanazawa, Ishikawa, Japan. .,Matrix Biology Department, the Kennedy Institute of Rheumatology Division, Imperial College London, Hammersmith, London, UK.
| | - Phoung Vo
- Matrix Biology Department, the Kennedy Institute of Rheumatology Division, Imperial College London, Hammersmith, London, UK
| | - Ioannis Kanakis
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, Liverpool, UK
| | - Ke Liu
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, Liverpool, UK
| | - George Bou-Gharios
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, Liverpool, UK
| |
Collapse
|
21
|
Proença S, Antunes B, Guedes RC, Ramilo-Gomes F, Cabral MF, Costa J, Fernandes AS, Castro M, Oliveira NG, Miranda JP. Pyridine-Containing Macrocycles Display MMP-2/9 Inhibitory Activity and Distinct Effects on Migration and Invasion of 2D and 3D Breast Cancer Models. Int J Mol Sci 2019; 20:E5109. [PMID: 31618886 PMCID: PMC6829403 DOI: 10.3390/ijms20205109] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/01/2019] [Accepted: 10/09/2019] [Indexed: 12/20/2022] Open
Abstract
The role of metalloproteinases (MMPs) on the migration and invasion of cancer cells has been correlated with tumor aggressiveness, namely with the up-regulation of MMP-2 and 9. Herein, two pyridine-containing macrocyclic compounds, [15]pyN5 and [16]pyN5, were synthesized, chemically characterized and evaluated as potential MMP inhibitors for breast cancer therapy using 3D and 2D cellular models. [15]pyN5 and [16]pyN5 (5-20 µM) showed a marked inhibition of MMPs activity (100% at concentrations ≥ 7.5 μM) when compared to ARP-100, a known MMP inhibitor. The inhibitory activity of [15]pyN5 and [16]pyN5 was further supported through in silico docking studies using Goldscore and ChemPLP scoring functions. Moreover, although no significant differences were observed in the invasion studies in the presence of all MMPs inhibitors, cell migration was significantly inhibited by both pyridine-containing macrocycles at concentrations above 5 μM in 2D cells (p < 0.05). In spheroids, the same effect was observed, but only with [16]pyN5 at 20 μM and ARP-100 at 40 μM. Overall, [15]pyN5 and [16]pyN5 led to impaired breast cancer cell migration and revealed to be potential inhibitors of MMPs 2 and 9.
Collapse
Affiliation(s)
- Susana Proença
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal.
- Institute for Risk Assessment Sciences, Utrecht University, P.O. Box 80177, 3508TD Utrecht, The Netherlands.
| | - Bernardo Antunes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal.
| | - Rita C Guedes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal.
| | - Filipa Ramilo-Gomes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal.
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisbon, Portugal.
| | - M Fátima Cabral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal.
| | - Judite Costa
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal.
| | | | - Matilde Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal.
| | - Nuno G Oliveira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal.
| | - Joana P Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal.
| |
Collapse
|
22
|
Cho CH, Lho YM, Hwang I, Kim DH. Role of matrix metalloproteinases 2 and 9 in the development of frozen shoulder: human data and experimental analysis in a rat contracture model. J Shoulder Elbow Surg 2019; 28:1265-1272. [PMID: 30846222 DOI: 10.1016/j.jse.2018.11.072] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 02/01/2023]
Abstract
BACKGROUND Although frozen shoulder (FS) is a common shoulder disorder, its pathogenesis is not yet determined. The function of matrix metalloproteinases (MMPs) is related to extracellular matrix remodeling. The purposes of this study were to investigate the pattern of sequential expression of MMPs in a rat model of shoulder contracture and to compare the expression of MMPs in the joint capsule between patients with FS and a control group. METHODS We obtained joint capsules from rats immobilized by molding plaster (a shoulder contracture model) at baseline, 3 days, 1 week, and 3 weeks (4 rats per time point; 16 rats in total). The expression of the inflammatory cytokine interleukin 6 (IL-6), MMP-2, and MMP-9 was examined by immunohistochemistry. We also obtained joint capsules from 21 patients with FS and 13 control patients with instability to quantify the expression levels of MMP-2 and MMP-9 by immunohistochemistry. RESULTS In the rat model, IL-6 and MMP-9 tended to be overexpressed in the joint capsule at 3 days and 1 week and MMP-2 at 3 days, 1 week, and 3 weeks. MMP-2 and MMP-9 were significantly overexpressed in the joint capsules of the patients with FS compared with those of control patients. CONCLUSION The results from both human and animal studies suggest the involvement of MMP-2 and MMP-9 in the development of FS. Animal study showed that the sequential expression of IL-6 and MMPs may be associated with fibrosis of the joint capsule.
Collapse
Affiliation(s)
- Chul-Hyun Cho
- Department of Orthopedic Surgery, Dongsan Medical Center, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Yun-Mee Lho
- Department of Biochemistry, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Ilseon Hwang
- Department of Pathology, Dongsan Medical Center, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Du Hwan Kim
- Department of Rehabilitation Medicine, Dongsan Medical Center, School of Medicine, Keimyung University, Daegu, Republic of Korea.
| |
Collapse
|
23
|
Fischer T, Senn N, Riedl R. Design and Structural Evolution of Matrix Metalloproteinase Inhibitors. Chemistry 2019; 25:7960-7980. [DOI: 10.1002/chem.201805361] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 01/09/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Thomas Fischer
- Center of Organic and Medicinal Chemistry, Institute of Chemistry, and BiotechnologyZurich University of Applied Sciences (ZHAW) Einsiedlerstrasse 31 8820 Wädenswil Switzerland
| | - Nicole Senn
- Center of Organic and Medicinal Chemistry, Institute of Chemistry, and BiotechnologyZurich University of Applied Sciences (ZHAW) Einsiedlerstrasse 31 8820 Wädenswil Switzerland
| | - Rainer Riedl
- Center of Organic and Medicinal Chemistry, Institute of Chemistry, and BiotechnologyZurich University of Applied Sciences (ZHAW) Einsiedlerstrasse 31 8820 Wädenswil Switzerland
| |
Collapse
|
24
|
Lopez C, Park S, Edwards S, Vong S, Hou S, Lee M, Sauerland H, Lee JJ, Jeong KJ. Matrix Metalloproteinase-Deactivating Contact Lens for Corneal Melting. ACS Biomater Sci Eng 2019; 5:1195-1199. [PMID: 31692998 DOI: 10.1021/acsbiomaterials.8b01404] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Corneal melting is an uncontrolled, excessive degradation of cellular and extracellular components of the cornea. This potential cause of corneal blindness is caused by excessive expression of zinc-dependent matrix metalloproteinases (MMPs) and has no satisfying cure as of now. Herein, we introduce a novel therapeutic hydrogel which can be made into a contact lens to slow down the progression of corneal melting by deactivating MMPs. The hydrogel backbone is comprised of poly(2-hydroxyetyl methacrylate) (pHEMA), a main material for commercial contact lenses, and dipicolylamine (DPA) which has high affinity and selectivity towards zinc ion. Due to the high affinity towards zinc ions, the DPA-conjugated pHEMA (pDPA-HEMA) hydrogel selectively removes zinc ions from a physiological buffer and deactivates MMP-1, MMP-2 and MMP-9 within 2 hours. pDPA-HEMA hydrogel also effectively prevents degradation of porcine corneas by collagenase A, a zinc-dependent protease, whereas the corneas completely degrades within 15 hours when incubated with pHEMA hydrogel. The presence of pDPA-HEMA hydrogel does not affect the viability of keratocytes and corneal epithelial cells. Unlike the conventional MMP inhibitors (MMPi), the pDPA-HEMA hydrogel minimizes the risk of serious non-specific side effects, and provides a method to slow down the progression of corneal melting and other related ocular diseases.
Collapse
Affiliation(s)
- Chelsi Lopez
- Department of Chemistry, University of Colorado Denver, Denver, CO 80204
| | - Shiwha Park
- Department of Chemical Engineering, University of New Hampshire, Durham, NH 03824
| | - Seth Edwards
- Department of Chemical Engineering, University of New Hampshire, Durham, NH 03824
| | - Selina Vong
- Department of Chemistry, University of Colorado Denver, Denver, CO 80204
| | - Shujie Hou
- Department of Chemical Engineering, University of New Hampshire, Durham, NH 03824
| | - Minyoung Lee
- Department of Chemistry, University of Colorado Denver, Denver, CO 80204
| | - Hunter Sauerland
- Department of Chemistry, University of Colorado Denver, Denver, CO 80204
| | - Jung-Jae Lee
- Department of Chemistry, University of Colorado Denver, Denver, CO 80204.,Department of Bioengineering, University of Colorado Denver, Aurora, CO 80045
| | - Kyung Jae Jeong
- Department of Chemical Engineering, University of New Hampshire, Durham, NH 03824
| |
Collapse
|
25
|
Myasoedova VA, Chistiakov DA, Grechko AV, Orekhov AN. Matrix metalloproteinases in pro-atherosclerotic arterial remodeling. J Mol Cell Cardiol 2018; 123:159-167. [PMID: 30172754 DOI: 10.1016/j.yjmcc.2018.08.026] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 08/07/2018] [Accepted: 08/29/2018] [Indexed: 12/20/2022]
Abstract
Matrix metalloproteinases (MMPs) is a family of Zn2+ endopeptidases that process various components of the extracellular matrix. These enzymes are also involved in activation and inhibition of signaling cascades through proteolytic cleavage of surface receptors. Moreover, MMPs play a key role in tissue remodeling and and repair. Dysregulation of MMPs is observed in patholofgical conditions, including atherosclerosis, which is associated with hyperactivation of MMPs, aberrant tissue remodeling and neovascularization of the growing atherosclerotic plaques. This makes MMPs interesting therapeutic targets that can be employed for developing novel therapies to treat atherosclerosis and its complications. Currently, a growing number of synthetic MMP inhibitors is available. In this review, we will discuss the role of these enzymes in atherosclerosis pathology and the ways of their pothential therapeutic use.
Collapse
Affiliation(s)
- Veronika A Myasoedova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | - Dimitry A Chistiakov
- Department of Basic and Applied Neurobiology, Serbsky Federal Medical Research Center for Psychiatry and Narcology, 119991 Moscow, Russia
| | - Andrey V Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 109240 Moscow, Russia
| | - Alexander N Orekhov
- Department of Basic and Applied Neurobiology, Serbsky Federal Medical Research Center for Psychiatry and Narcology, 119991 Moscow, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia.
| |
Collapse
|
26
|
de Miguel I, Orbe J, Sánchez-Arias JA, Rodríguez JA, Salicio A, Rabal O, Belzunce M, Sáez E, Xu M, Wu W, Tan H, Ma H, Páramo JA, Oyarzabal J. Phenotypic Screening To Discover Novel Chemical Series as Efficient Antihemorrhagic Agents. ACS Med Chem Lett 2018; 9:428-433. [PMID: 29795754 DOI: 10.1021/acsmedchemlett.7b00549] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 04/16/2018] [Indexed: 12/24/2022] Open
Abstract
In an effort to find novel chemical series as antifibrinolytic agents, we explore α-phenylsulfonyl-α-spiropiperidines bearing different zinc-binding groups (ZBGs) to target those metalloproteinases involved in the fibrinolytic process: MMP3 and MMP10. Surprisingly, all these new chemical series were inactive against these metalloproteinases; however, several new molecules retained the antifibrinolytic activity in a phenotypic functional assay using thromboelastometry and human whole blood. Further optimization led to compound 38 as a potent antifibrinolytic agent in vivo, three times more efficacious than the current standard-of-care (tranexamic acid, TXA) at 300 times lower dose. Finally, in order to decipher the underlying mode-of-action leading to this phenotypic response, an affinity-based probe 39 was successfully designed to identify the target involved in this response: a potentially unknown mechanism-of-action in the fibrinolytic process.
Collapse
Affiliation(s)
| | - Josune Orbe
- CIBER Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | | | - José A. Rodríguez
- CIBER Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Agustina Salicio
- CIBER Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | | | | | | | - Musheng Xu
- WuXi Apptec (Tianjin) Co. Ltd., TEDA, No. 111 HuangHai Road, Fourth Avenue, Tianjin 300456, P. R. China
| | - Wei Wu
- WuXi Apptec (Tianjin) Co. Ltd., TEDA, No. 111 HuangHai Road, Fourth Avenue, Tianjin 300456, P. R. China
| | - Haizhong Tan
- WuXi Apptec (Tianjin) Co. Ltd., TEDA, No. 111 HuangHai Road, Fourth Avenue, Tianjin 300456, P. R. China
| | - Hongyu Ma
- WuXi Apptec (Tianjin) Co. Ltd., TEDA, No. 111 HuangHai Road, Fourth Avenue, Tianjin 300456, P. R. China
| | - José A. Páramo
- Hematology Service, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | | |
Collapse
|
27
|
Jha T, Adhikari N, Saha A, Amin SA. Multiple molecular modelling studies on some derivatives and analogues of glutamic acid as matrix metalloproteinase-2 inhibitors. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2018; 29:43-68. [PMID: 29254380 DOI: 10.1080/1062936x.2017.1406986] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 11/15/2017] [Indexed: 06/07/2023]
Abstract
Matrix metalloproteinase-2 (MMP-2) is a potential target in anticancer drug discovery due to its association with angiogenesis, metastasis and tumour progression. In this study, 67 glutamic acid derivatives, synthesized and evaluated as MMP-2 inhibitors, were taken into account for multi-QSAR modelling study (regression-based 2D-QSAR, classification-based LDA-QSAR, Bayesian classification QSAR, HQSAR, 3D-QSAR CoMFA and CoMSIA as well as Open3DQSAR). All these QSAR studies were statistically validated individually. Regarding the 3D-QSAR analysis, the Open3DQSAR results were better than CoMFA and CoMSIA, although all these 3D-QSAR models supported each other. The importance of biphenylsulphonyl moiety over phenylacetyl/naphthylacetyl moieties was established due to its association with favourable steric and hydrophobic characters. HQSAR, LDA-QSAR and Bayesian classification QSAR studies also suggested that the biphenylsulphonamido group was better than the phenylacetylcarboxamido function. Additionally, glutamines were proven to be far better inhibitors than isoglutamines. Observations obtained from the current study were revalidated and supported by the earlier reported molecular modelling studies. Depending on these observations, newer glutamic acid-based compounds may be designed further in future for potent MMP-2 inhibitory activity.
Collapse
Affiliation(s)
- T Jha
- a Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry , Department of Pharmaceutical Technology , Jadavpur University , Kolkata , India
| | - N Adhikari
- a Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry , Department of Pharmaceutical Technology , Jadavpur University , Kolkata , India
| | - A Saha
- b Department of Chemical Technology , University of Calcutta , Kolkata , India
| | - S A Amin
- a Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry , Department of Pharmaceutical Technology , Jadavpur University , Kolkata , India
| |
Collapse
|
28
|
Wang X, Khalil RA. Matrix Metalloproteinases, Vascular Remodeling, and Vascular Disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 81:241-330. [PMID: 29310800 DOI: 10.1016/bs.apha.2017.08.002] [Citation(s) in RCA: 421] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases that degrade various proteins in the extracellular matrix (ECM). Typically, MMPs have a propeptide sequence, a catalytic metalloproteinase domain with catalytic zinc, a hinge region or linker peptide, and a hemopexin domain. MMPs are commonly classified on the basis of their substrates and the organization of their structural domains into collagenases, gelatinases, stromelysins, matrilysins, membrane-type (MT)-MMPs, and other MMPs. MMPs are secreted by many cells including fibroblasts, vascular smooth muscle (VSM), and leukocytes. MMPs are regulated at the level of mRNA expression and by activation through removal of the propeptide domain from their latent zymogen form. MMPs are often secreted in an inactive proMMP form, which is cleaved to the active form by various proteinases including other MMPs. MMPs degrade various protein substrates in ECM including collagen and elastin. MMPs could also influence endothelial cell function as well as VSM cell migration, proliferation, Ca2+ signaling, and contraction. MMPs play a role in vascular tissue remodeling during various biological processes such as angiogenesis, embryogenesis, morphogenesis, and wound repair. Alterations in specific MMPs could influence arterial remodeling and lead to various pathological disorders such as hypertension, preeclampsia, atherosclerosis, aneurysm formation, as well as excessive venous dilation and lower extremity venous disease. MMPs are often regulated by endogenous tissue inhibitors of metalloproteinases (TIMPs), and the MMP/TIMP ratio often determines the extent of ECM protein degradation and tissue remodeling. MMPs may serve as biomarkers and potential therapeutic targets for certain vascular disorders.
Collapse
Affiliation(s)
- Xi Wang
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
29
|
Levine JM, Cohen ND, Fandel TM, Levine GJ, Mankin J, Griffin JF, Kerwin SC, Boudreau CE, Trivedi A, Noble-Haeusslein LJ. Early Blockade of Matrix Metalloproteinases in Spinal-Cord–Injured Dogs Results in a Long-Term Increase in Bladder Compliance. J Neurotrauma 2017; 34:2656-2667. [DOI: 10.1089/neu.2017.5001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Jonathan M. Levine
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Noah D. Cohen
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Thomas M. Fandel
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Gwendolyn J. Levine
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Joseph Mankin
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - John F. Griffin
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Sharon C. Kerwin
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - C. Elizabeth Boudreau
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Alpa Trivedi
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Linda J. Noble-Haeusslein
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| |
Collapse
|
30
|
Gayan S, Teli A, Dey T. Inherent aggressive character of invasive and non-invasive cells dictates the in vitro migration pattern of multicellular spheroid. Sci Rep 2017; 7:11527. [PMID: 28912559 PMCID: PMC5599661 DOI: 10.1038/s41598-017-10078-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 07/21/2017] [Indexed: 11/24/2022] Open
Abstract
Cellular migration, a process relevant to metastasis, is mostly studied in the conventional 2D condition. However, cells cultured in the 3D condition assumed to mimic the in vivo conditions better. The current study is designed to compare an invasive and non-invasive adenocarcinoma cell with an invasive fibrosarcoma cell to understand the migration pattern of the multicellular spheroid. It is observed that conventional haplotaxis, chemotactic and pseudo-3D migration assay cannot distinguish between the invasive and non-invasive cells conclusively under 2D condition. Invasive spheroids migrate rapidly in sprouting assay in comparison to non-invasive spheroids. Effects of cytochalasin B, marimastat and blebbistatin are tested to determine the influence of different migration modality namely actin polymerization, matrix metalloprotease and acto-myosin in both culture conditions. Altered mRNA profile of cellular migration related genes (FAK, Talin, Paxillin, p130cas and Vinculin) is observed between 2D and 3D condition followed by the changed expression of matrix metallo proteases. A distinct difference is observed in distribution and formation of focal adhesion complex under these culture conditions. This study demonstrates the efficacy of multicellular spheroids in identifying the intrinsic aggressive behavior of different cell lines as a proof of concept and recognizes the potential of spheroids as a migration model.
Collapse
Affiliation(s)
- Sukanya Gayan
- Institute of Bioinformatics and Biotechnology,Savitribai Phule Pune University, Pune, India
| | - Abhishek Teli
- Institute of Bioinformatics and Biotechnology,Savitribai Phule Pune University, Pune, India
| | - Tuli Dey
- Institute of Bioinformatics and Biotechnology,Savitribai Phule Pune University, Pune, India.
| |
Collapse
|
31
|
Development of a Non-Hydroxamate Dual Matrix Metalloproteinase (MMP)-7/-13 Inhibitor. Molecules 2017; 22:molecules22091548. [PMID: 32961647 PMCID: PMC6151531 DOI: 10.3390/molecules22091548] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 09/08/2017] [Accepted: 09/08/2017] [Indexed: 12/12/2022] Open
Abstract
Matrix metalloproteinase 7 (MMP-7) is a member of the MMP superfamily and is able to degrade extracellular matrix proteins such as casein, gelatin, fibronectin and proteoglycan. MMP-7 is a validated target for the development of small molecule drugs against cancer. MMP-13 is within the enzyme class the most efficient contributor to type II collagen degeneration and is a validated target in arthritis and cancer. We have developed the dual MMP-7/-13 inhibitor ZHAWOC6941 with IC50-values of 2.2 μM (MMP-7) and 1.2 μM (MMP-13) that is selective over a broad range of MMP isoforms. It spares MMP-1, -2, -3, -8, -9, -12 and -14, making it a valuable modulator for targeted polypharmacology approaches.
Collapse
|
32
|
Selective matrix metalloproteinase inhibition increases breaking strength and reduces anastomotic leakage in experimentally obstructed colon. Int J Colorectal Dis 2017; 32:1277-1284. [PMID: 28717842 DOI: 10.1007/s00384-017-2857-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/07/2017] [Indexed: 02/04/2023]
Abstract
PURPOSE Colonic obstruction causes loss of collagen and impairment of anastomotic integrity by matrix metalloproteinases (MMPs). Unexpectedly, pharmacological MMP inhibition increased anastomotic leakage (AL) in obstructed colon possibly due to the non-selective nature of these compounds and the experimental model applied. We therefore studied the effects of selective MMP inhibition on the healing of anastomoses in colon obstructed by a novel laparoscopic technique. METHODS Left colon was obstructed in 38 male Sprague-Dawley rats (226-284 g). After 12 h, stenoses were resected and end-to-end anastomoses constructed. Baseline breaking strength was determined in 6 animals on day 0. The remaining 32 rats were randomized to daily treatment with the selective MMP-8, MMP-9, and MMP-12 inhibitor AZD3342 (n = 16) or vehicle (n = 16). On day 3, anastomoses were evaluated for AL and breaking strength. Isolated anastomotic wound tissue was analyzed on total collagen and pepsin-insoluble and pepsin-soluble collagen by hydroxyproline. The soluble collagens were further differentiated into native, measured by Sircol, and fragmented forms. RESULTS Baseline breaking strength was maintained with AZD3342 but decreased by 25% (P = 0.023) in the vehicle group. The anastomotic breaking strength of AZD3342-treated rats was 44% higher (P = 0.008) than the vehicle-treated rats. Furthermore, the AL rate was reduced (P = 0.037) with AZD3342 compared with vehicle treatment. AZD3342 treatment influenced neither the total or insoluble collagen concentrations nor the degree of fragmentation of the soluble collagen triple helices. CONCLUSION Selective MMP inhibition increased anastomotic breaking strength and reduced AL after resection of colonic obstruction.
Collapse
|
33
|
Abstract
Most toxic physeal changes are characterized microscopically by altered chondrocyte development, proliferation, or maturation in the growth plate and eventually result in disordered appositional bone growth. Many therapeutic drugs directly or indirectly target proteins involved in chondrocytic differentiation and maturation pathways, so toxic physeal injury has become increasingly common in preclinical toxicologic pathology. While physeal dysplasia has been associated with several different drug classes including bisphosphonates, vascular endothelial growth factor receptor inhibitors, fibroblast growth factor receptor inhibitors, transforming growth factor beta receptor inhibitors, and vascular targeting agents, physeal changes often share similar morphologic features including thickening and disorganization of the hypertrophic layer, increased numbers of hypertrophic chondrocytes, altered mineralization of endochondral ossification, and/or increased thickness of subphyseal bone. Knowledge of genetic and nutritional diseases affecting bone growth has been important in helping to determine which specific target drugs may be affecting that could result in toxic physeal lesions. A pathophysiologic mechanism for most physeal toxicants has been determined in detail using a variety of investigative techniques. However, due to the signaling cross talk and the tight regulation required for chondrocyte maturation in the physis, several growth factor pathways are likely to be affected simultaneously with pharmacologic disruption of physeal homeostasis and inhibition of one factor necessary for chondrocyte function often affects others.
Collapse
|
34
|
Abstract
Toxic tendinopathy is a rare but reproducible complication in humans, given agents of four drug classes: aromatase inhibitors, fluoroquinolone antibiotics, glucocorticoids (long-term regimens), and statins. Toxic tendinopathy in humans has been linked less consistently to treatment with anabolic steroids, antiretroviral agents (mainly protease inhibitors), metalloproteinase inhibitors (MMPI), and isotretinoin. Classic drug-induced tendinopathies appear as "tendinosis" (i.e., progressive tendon degeneration without inflammation), although cases associated with aromatase inhibitors exhibit mainly tenosynovitis. Any tendon may be affected, but fluoroquinolones, glucocorticoids, and statins most frequently affect large load-bearing tendons in the lower limb, especially the calcaneal ("Achilles") tendon-which ruptures in approximately 30 to 40% of cases. The time to symptom onset ranges from days (fluoroquinolones) to weeks, months, or even years. The pathogenesis is incompletely understood, but proposed mechanisms include apoptosis of tenoblasts and tenocytes, deficient tenocyte function (leading to abnormal extracellular matrix maintenance and repair as well as disrupted intercellular signaling), and structural disintegration (via a combination of increased expression of lytic enzymes, lessened cholesterol content in cell membranes, and neoangiogenesis within highly ordered tendon tissue). Nonclinical safety assessment of therapeutic candidates in these drug classes should incorporate tendon routinely as a protocol-specified tissue for pathology evaluation.
Collapse
Affiliation(s)
- Brad Bolon
- 1 GEMpath, Inc., Longmont, Colorado, USA
| |
Collapse
|
35
|
Matrix Metalloproteinase Inhibitors as Investigational and Therapeutic Tools in Unrestrained Tissue Remodeling and Pathological Disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 148:355-420. [PMID: 28662828 DOI: 10.1016/bs.pmbts.2017.04.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Matrix metalloproteinases (MMPs) are zinc-dependent proteolytic enzymes that degrade various proteins in the extracellular matrix (ECM). MMPs may also regulate the activity of membrane receptors and postreceptor signaling mechanisms and thereby affect cell function. The MMP family includes collagenases, gelatinases, stromelysins, matrilysins, membrane-type MMPs, and other MMPs. Inactive proMMPs are cleaved by other MMPs or proteases into active MMPs, which interact with various protein substrates in ECM and cell surface. MMPs regulate important biological processes such as vascular remodeling and angiogenesis and may be involved in the pathogenesis of cardiovascular disorders such as hypertension, atherosclerosis, and aneurysm. The role of MMPs is often assessed by measuring their mRNA expression, protein levels, and proteolytic activity using gel zymography. MMP inhibitors are also used to assess the role of MMPs in different biological processes and pathological conditions. MMP activity is regulated by endogenous tissue inhibitors of metalloproteinases (TIMPs), and the MMP/TIMP balance could determine the net MMP activity, ECM turnover, and tissue remodeling. Also, several synthetic MMP inhibitors have been developed. Synthetic MMP inhibitors include a large number of zinc-binding globulins (ZBGs), in addition to non-ZBGs and mechanism-based inhibitors. MMP inhibitors have been proposed as potential tools in the management of osteoarthritis, cancer, and cardiovascular disorders. However, most MMP inhibitors have broad-spectrum actions on multiple MMPs and could cause undesirable musculoskeletal side effects. Currently, doxycycline is the only MMP inhibitor approved by the Food and Drug Administration. New generation biological and synthetic MMP inhibitors may show greater MMP specificity and fewer side effects and could be useful in targeting specific MMPs, reducing unrestrained tissue remodeling, and the management of MMP-related pathological disorders.
Collapse
|
36
|
Chen Y, Peng W, Raffetto JD, Khalil RA. Matrix Metalloproteinases in Remodeling of Lower Extremity Veins and Chronic Venous Disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 147:267-299. [PMID: 28413031 DOI: 10.1016/bs.pmbts.2017.02.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The veins of the lower extremity are equipped with efficient wall, contractile vascular smooth muscle (VSM), and competent valves in order to withstand the high venous hydrostatic pressure in the lower limb and allow unidirectional movement of deoxygenated blood toward the heart. The vein wall structure and function are in part regulated by matrix metalloproteinases (MMPs). MMPs are zinc-dependent endopeptidases that are secreted as inactive pro-MMPs by different cells in the venous wall including fibroblasts, VSM, and leukocytes. Pro-MMPs are activated by other MMPs, proteinases, and other endogenous and exogenous activators. MMPs degrade various extracellular matrix (ECM) proteins including collagen and elastin, and could affect other cellular processes including endothelium-mediated dilation, VSM cell migration, and proliferation as well as modulation of Ca2+ signaling and contraction in VSM. It is thought that increased lower limb venous hydrostatic pressure increases hypoxia-inducible factors and other MMP inducers such as extracellular matrix metalloproteinase inducer, leading to increased MMP expression/activity, ECM protein degradation, vein wall relaxation, and venous dilation. Vein wall inflammation and leukocyte infiltration cause additional increases in MMPs, and further vein wall dilation and valve degradation, that could lead to chronic venous disease and varicose veins (VVs). VVs are often presented as vein wall dilation and tortuosity, incompetent venous valves, and venous reflux. Different regions of VVs show different MMP levels and ECM proteins with atrophic regions showing high MMP levels/activity and little ECM compared to hypertrophic regions with little or inactive MMPs and abundant ECM. Treatment of VVs includes compression stockings, venotonics, sclerotherapy, or surgical removal. However, these approaches do not treat the cause of VVs, and other lines of treatment may be needed. Modulation of endogenous tissue inhibitors of metalloproteinases (TIMPs), and exogenous synthetic MMP inhibitors may provide new approaches in the management of VVs.
Collapse
Affiliation(s)
- Yunfei Chen
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Wei Peng
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Joseph D Raffetto
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
37
|
Arylsulfonamides and selectivity of matrix metalloproteinase-2: An overview. Eur J Med Chem 2017; 129:72-109. [DOI: 10.1016/j.ejmech.2017.02.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 02/02/2017] [Accepted: 02/06/2017] [Indexed: 12/14/2022]
|
38
|
Isaacson KJ, Martin Jensen M, Subrahmanyam NB, Ghandehari H. Matrix-metalloproteinases as targets for controlled delivery in cancer: An analysis of upregulation and expression. J Control Release 2017; 259:62-75. [PMID: 28153760 DOI: 10.1016/j.jconrel.2017.01.034] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/18/2017] [Accepted: 01/26/2017] [Indexed: 02/07/2023]
Abstract
While commonly known for degradation of the extracellular matrix, matrix metalloproteinases (MMPs) exhibit broad potential for use in targeting of bioactive and imaging agents in cancer treatment. MMPs are upregulated at all stages of expression in cancers. A comprehensive analysis of published literature on expression of all MMP subtypes at the genetic, protein, and activity levels in normal and diseased tissues indicate targeting applicability in a variety of cancers. This expression significantly increases at advanced cancer stages, providing an improved opportunity for controlled release in higher-stage patients. Since MMPs are integral at every stage of metastasis, MMP roles in cancer are discussed with a focus on MMP distribution and mobility within cells and tumors for cancer targeting applications. Several strategies for MMP utilization in targeting - such as matrix degradation, MMP cleavage, MMP binding, and MMP-induced environmental changes - are addressed.
Collapse
Affiliation(s)
- Kyle J Isaacson
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - M Martin Jensen
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - Nithya B Subrahmanyam
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - Hamidreza Ghandehari
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
39
|
Fischer T, Riedl R. Targeted Fluoro Positioning for the Discovery of a Potent and Highly Selective Matrix Metalloproteinase Inhibitor. ChemistryOpen 2017; 6:192-195. [PMID: 28413749 PMCID: PMC5390795 DOI: 10.1002/open.201600158] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Indexed: 12/28/2022] Open
Abstract
The incorporation of fluorine atoms into functional molecules is of wide interest in synthetic organic chemistry as well as cognate disciplines. In particular, in medicinal chemistry, there is a strong desire to positively influence the physicochemical molecular properties of drug compounds by introducing fluorine into biologically active molecules. Here, we present targeted fluoro positioning as the key design principle of converting a weak matrix metalloproteinase‐13 (MMP‐13) inhibitor into a very potent (IC50=6 nm) and highly selective (selectivity factors of >1000 over MMP‐1, 2, 3, 7, 8, 9, 10, 12, 14) inhibitor with excellent plasma and microsomal stability, and no binding to the hERG channel (hERG: human ether‐a‐go‐go related gene).
Collapse
Affiliation(s)
- Thomas Fischer
- Institute of Chemistry and Biotechnology Center for Organic and Medicinal Chemistry Zurich University of Applied Sciences (ZHAW) Einsiedlerstrasse 318820 Wädenswil Switzerland
| | - Rainer Riedl
- Institute of Chemistry and Biotechnology Center for Organic and Medicinal Chemistry Zurich University of Applied Sciences (ZHAW) Einsiedlerstrasse 318820 Wädenswil Switzerland
| |
Collapse
|
40
|
Muvva C, Patra S, Venkatesan S. MMpI: A WideRange of Available Compounds of Matrix Metalloproteinase Inhibitors. PLoS One 2016; 11:e0159321. [PMID: 27509041 PMCID: PMC4979873 DOI: 10.1371/journal.pone.0159321] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 06/30/2016] [Indexed: 11/19/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent proteinases involved in the regulation of the extracellular signaling and structural matrix environment of cells and tissues. MMPs are considered as promising targets for the treatment of many diseases. Therefore, creation of database on the inhibitors of MMP would definitely accelerate the research activities in this area due to its implication in above-mentioned diseases and associated limitations in the first and second generation inhibitors. In this communication, we report the development of a new MMpI database which provides resourceful information for all researchers working in this field. It is a web-accessible, unique resource that contains detailed information on the inhibitors of MMP including small molecules, peptides and MMP Drug Leads. The database contains entries of ~3000 inhibitors including ~72 MMP Drug Leads and ~73 peptide based inhibitors. This database provides the detailed molecular and structural details which are necessary for the drug discovery and development. The MMpI database contains physical properties, 2D and 3D structures (mol2 and pdb format files) of inhibitors of MMP. Other data fields are hyperlinked to PubChem, ChEMBL, BindingDB, DrugBank, PDB, MEROPS and PubMed. The database has extensive searching facility with MMpI ID, IUPAC name, chemical structure and with the title of research article. The MMP inhibitors provided in MMpI database are optimized using Python-based Hierarchical Environment for Integrated Xtallography (Phenix) software. MMpI Database is unique and it is the only public database that contains and provides the complete information on the inhibitors of MMP. Database URL: http://clri.res.in/subramanian/databases/mmpi/index.php.
Collapse
Affiliation(s)
- Charuvaka Muvva
- Chemical Laboratory, Council of Scientific and Industrial Research-Central Leather Research Institute, Chennai, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Sanjukta Patra
- Department of Biotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Subramanian Venkatesan
- Chemical Laboratory, Council of Scientific and Industrial Research-Central Leather Research Institute, Chennai, India
- * E-mail: ;
| |
Collapse
|
41
|
Fossey S, Vahle J, Long P, Schelling S, Ernst H, Boyce RW, Jolette J, Bolon B, Bendele A, Rinke M, Healy L, High W, Roth DR, Boyle M, Leininger J. Nonproliferative and Proliferative Lesions of the Rat and Mouse Skeletal Tissues (Bones, Joints, and Teeth). J Toxicol Pathol 2016; 29:49S-103S. [PMID: 27621538 PMCID: PMC5013709 DOI: 10.1293/tox.29.3s-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The INHAND (International Harmonization of Nomenclature and Diagnostic Criteria for Lesions in Rats and Mice) Project (www.toxpath.org/inhand.asp) is an initiative of the Societies of Toxicological Pathology from Europe (ESTP), Great Britain (BSTP), Japan (JSTP) and North America (STP) to develop an internationally accepted nomenclature for proliferative and nonproliferative lesions in laboratory animals. The purpose of this publication is to provide a standardized nomenclature for classifying microscopic lesions observed in the skeletal tissues and teeth of laboratory rats and mice, with color photomicrographs illustrating examples of many common lesions. The standardized nomenclature presented in this document is also available on the internet (http://www.goreni.org/). Sources of material were databases from government, academic and industrial laboratories throughout the world.
Collapse
Affiliation(s)
| | - John Vahle
- Lilly Research Laboratories, Indianapolis, IN, USA
| | | | - Scott Schelling
- Pfizer Inc., Andover, MA, USA
- Dr. Schelling retired April 2015
| | | | | | | | | | | | | | - Laura Healy
- LNH Tox Path Consulting, LLC, Kalamazoo, MI, USA
| | - Wanda High
- WB High Preclin Path/Tox Consulting, LLC, Rochester, NY,
USA
| | | | | | - Joel Leininger
- JRL Consulting, LLC, Chapel Hill, NC, USA
- Chair of the Skeletal Tissues INHAND Committee
| |
Collapse
|
42
|
Dreymueller D, Ludwig A. Considerations on inhibition approaches for proinflammatory functions of ADAM proteases. Platelets 2016; 28:354-361. [PMID: 27460023 DOI: 10.1080/09537104.2016.1203396] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Proteases of the disintegrin and metalloproteinase (ADAM) family mediate the proteolytic shedding of various surface molecules including cytokine precursors, adhesion molecules, growth factors, and receptors. Within the vasculature ADAM10 and ADAM17 regulate endothelial permeability, transendothelial leukocyte migration, and the adhesion of leukocytes and platelets. In vivo studies show that both proteases are implicated in several inflammatory pathologies, for example, edema formation, leukocyte infiltration, and thrombosis. However, both proteases also contribute to developmental and regenerative processes. Thus, although ADAMs can be regarded as valuable drug targets in many aspects, the danger of severe side effects is clearly visible. To circumvent these side effects, traditional inhibition approaches have to be improved to target ADAMs at the right time in the right place. Moreover, the inhibitors need to be more selective for the target protease and if possible also for the substrate. Antibodies recognizing the active conformation of ADAMs or small molecules blocking exosites of ADAM proteases may represent inhibitors with the desired selectivities.
Collapse
Affiliation(s)
- Daniela Dreymueller
- a Institute of Pharmacology and Toxicology , RWTH Aachen University , Aachen , Germany
| | - Andreas Ludwig
- a Institute of Pharmacology and Toxicology , RWTH Aachen University , Aachen , Germany
| |
Collapse
|
43
|
Pavan M, Galesso D, Secchieri C, Guarise C. Hyaluronic acid alkyl derivative: A novel inhibitor of metalloproteases and hyaluronidases. Int J Biol Macromol 2016; 84:221-6. [PMID: 26709144 DOI: 10.1016/j.ijbiomac.2015.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 12/01/2015] [Indexed: 12/18/2022]
|
44
|
MacColl E, Khalil RA. Matrix Metalloproteinases as Regulators of Vein Structure and Function: Implications in Chronic Venous Disease. J Pharmacol Exp Ther 2015; 355:410-28. [PMID: 26319699 PMCID: PMC4658486 DOI: 10.1124/jpet.115.227330] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/27/2015] [Indexed: 12/24/2022] Open
Abstract
Lower-extremity veins have efficient wall structure and function and competent valves that permit upward movement of deoxygenated blood toward the heart against hydrostatic venous pressure. Matrix metalloproteinases (MMPs) play an important role in maintaining vein wall structure and function. MMPs are zinc-binding endopeptidases secreted as inactive pro-MMPs by fibroblasts, vascular smooth muscle (VSM), and leukocytes. Pro-MMPs are activated by various activators including other MMPs and proteinases. MMPs cause degradation of extracellular matrix (ECM) proteins such as collagen and elastin, and could have additional effects on the endothelium, as well as VSM cell migration, proliferation, Ca(2+) signaling, and contraction. Increased lower-extremity hydrostatic venous pressure is thought to induce hypoxia-inducible factors and other MMP inducers/activators such as extracellular matrix metalloproteinase inducer, prostanoids, chymase, and hormones, leading to increased MMP expression/activity, ECM degradation, VSM relaxation, and venous dilation. Leukocyte infiltration and inflammation of the vein wall cause further increases in MMPs, vein wall dilation, valve degradation, and different clinical stages of chronic venous disease (CVD), including varicose veins (VVs). VVs are characterized by ECM imbalance, incompetent valves, venous reflux, wall dilation, and tortuosity. VVs often show increased MMP levels, but may show no change or decreased levels, depending on the VV region (atrophic regions with little ECM versus hypertrophic regions with abundant ECM) and MMP form (inactive pro-MMP versus active MMP). Management of VVs includes compression stockings, venotonics, and surgical obliteration or removal. Because these approaches do not treat the causes of VVs, alternative methods are being developed. In addition to endogenous tissue inhibitors of MMPs, synthetic MMP inhibitors have been developed, and their effects in the treatment of VVs need to be examined.
Collapse
Affiliation(s)
- Elisabeth MacColl
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
45
|
Abstract
Animal models of osteoarthritis are extensively used for investigating disease pathways and for preclinical testing of novel therapies. Their predictive utility, however, has often been questioned, mainly because preclinical efficacy of novel therapeutics is poorly translated in clinical trials. In the current narrative review, we consider the preclinical models that were used to support undertaking clinical trials for disease-modifying osteoarthritis drugs, and compare outcomes between clinical and preclinical studies. We discuss this in light of the 1999 Food and Drug Administration draft guidelines for industry for use in the development of drugs, devices, and biological products intended for the treatment of osteoarthritis, which raised five considerations on the usefulness of osteoarthritis models. We systematically discuss what has been learnt regarding these five points since 1999, with emphasis on replicating distinct risk factors and subtypes of human osteoarthritis, and on comprehensive evaluation of the disease in animals, including pathology of all joint tissues, biomarker analysis, and assessment of pain and joint function. Finally, we discuss lessons learnt and propose some recommendations for how the evidence from preclinical research might be strengthened with a view to improving success in clinical translation.
Collapse
Affiliation(s)
- Anne-Marie Malfait
- Department of Medicine, Division of Rheumatology, and Department of Biochemistry, Rush University Medical Center, Chicago, IL, 60612, USA.
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Institute of Bone and Joint Research, University of Sydney at Royal North Shore Hospital, St Leonards, NSW, 2065, Australia.
| |
Collapse
|
46
|
Ding Y, O’Keefe H, DeLorey JL, Israel DI, Messer JA, Chiu CH, Skinner SR, Matico RE, Murray-Thompson MF, Li F, Clark MA, Cuozzo JW, Arico-Muendel C, Morgan BA. Discovery of Potent and Selective Inhibitors for ADAMTS-4 through DNA-Encoded Library Technology (ELT). ACS Med Chem Lett 2015; 6:888-93. [PMID: 26288689 DOI: 10.1021/acsmedchemlett.5b00138] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 07/07/2015] [Indexed: 12/12/2022] Open
Abstract
The aggrecan degrading metalloprotease ADAMTS-4 has been identified as a novel therapeutic target for osteoarthritis. Here, we use DNA-encoded Library Technology (ELT) to identify novel ADAMTS-4 inhibitors from a DNA-encoded triazine library by affinity selection. Structure-activity relationship studies based on the selection information led to the identification of potent and highly selective inhibitors. For example, 4-(((4-(6,7-dimethoxy-3,4-dihydroisoquinolin-2(1H)-yl)-6-(((4-methylpiperazin-1-yl)methyl)amino)-1,3,5-triazin-2-yl)amino)methyl)-N-ethyl-N-(m-tolyl)benzamide has IC50 of 10 nM against ADAMTS-4, with >1000-fold selectivity over ADAMT-5, MMP-13, TACE, and ADAMTS-13. These inhibitors have no obvious zinc ligand functionality.
Collapse
Affiliation(s)
- Yun Ding
- Platform Technology & Science, GlaxoSmithKline, ELT-Boston, 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Heather O’Keefe
- Platform Technology & Science, GlaxoSmithKline, ELT-Boston, 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Jennifer L. DeLorey
- Tedor Pharma, Inc., 400 Highland Corporate Drive, Cumberland, Rhode Island 02864, United States
| | - David I. Israel
- Platform Technology & Science, GlaxoSmithKline, ELT-Boston, 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Jeffrey A. Messer
- Platform Technology & Science, GlaxoSmithKline, ELT-Boston, 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Cynthia H. Chiu
- Platform Technology & Science, GlaxoSmithKline, ELT-Boston, 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Steven R. Skinner
- Platform Technology & Science, GlaxoSmithKline, ELT-Boston, 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Rosalie E. Matico
- Biological
Reagent and Assay Development, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Monique F. Murray-Thompson
- Biological
Reagent and Assay Development, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Fan Li
- Tufts Healthcare Institute, 136 Harrison Avenue, Boston, Massachusetts 02111, United States
| | - Matthew A. Clark
- X-Chem, Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - John W. Cuozzo
- X-Chem, Inc., 100 Beaver Street, Waltham, Massachusetts 02453, United States
| | - Christopher Arico-Muendel
- Platform Technology & Science, GlaxoSmithKline, ELT-Boston, 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Barry A. Morgan
- Center for Drug Discovery, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| |
Collapse
|
47
|
Knapinska AM, Dreymuller D, Ludwig A, Smith L, Golubkov V, Sohail A, Fridman R, Giulianotti M, LaVoi TM, Houghten RA, Fields GB, Minond D. SAR Studies of Exosite-Binding Substrate-Selective Inhibitors of A Disintegrin And Metalloprotease 17 (ADAM17) and Application as Selective in Vitro Probes. J Med Chem 2015; 58:5808-24. [PMID: 26192023 DOI: 10.1021/acs.jmedchem.5b00354] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
ADAM17 is implicated in several debilitating diseases. However, drug discovery efforts targeting ADAM17 have failed due to the utilization of zinc-binding inhibitors. We previously reported discovery of highly selective nonzinc-binding exosite-targeting inhibitors of ADAM17 that exhibited not only enzyme isoform selectivity but synthetic substrate selectivity as well ( J. Biol. Chem. 2013, 288, 22871). As a result of SAR studies presented herein, we obtained several highly selective ADAM17 inhibitors, six of which were further characterized in biochemical and cell-based assays. Lead compounds exhibited low cellular toxicity and high potency and selectivity for ADAM17. In addition, several of the leads inhibited ADAM17 in a substrate-selective manner, which has not been previously documented for inhibitors of the ADAM family. These findings suggest that targeting exosites of ADAM17 can be used to obtain highly desirable substrate-selective inhibitors. Additionally, current inhibitors can be used as probes of biological activity of ADAM17 in various in vitro and, potentially, in vivo systems.
Collapse
Affiliation(s)
- Anna M Knapinska
- ∥Florida Atlantic University, 5353 Parkside Drive, Jupiter, Florida 33458, United States
| | - Daniela Dreymuller
- ⊥Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| | - Andreas Ludwig
- ⊥Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| | - Lyndsay Smith
- ∥Florida Atlantic University, 5353 Parkside Drive, Jupiter, Florida 33458, United States
| | - Vladislav Golubkov
- ‡Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Anjum Sohail
- §Wayne State University, 8200 Scott Hall, 540 East Canfield Avenue, Detroit, Michigan 48201, United States
| | - Rafael Fridman
- §Wayne State University, 8200 Scott Hall, 540 East Canfield Avenue, Detroit, Michigan 48201, United States
| | - Marc Giulianotti
- †Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States.,∇Department of Chemistry, Center for Drug Discovery and Innovation, University of South Florida, Tampa, Florida 33612, United States
| | - Travis M LaVoi
- †Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Richard A Houghten
- †Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Gregg B Fields
- ∥Florida Atlantic University, 5353 Parkside Drive, Jupiter, Florida 33458, United States.,#The Scripps Research Institute/Scripps Florida, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Dmitriy Minond
- †Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| |
Collapse
|
48
|
Marshall DC, Lyman SK, McCauley S, Kovalenko M, Spangler R, Liu C, Lee M, O’Sullivan C, Barry-Hamilton V, Ghermazien H, Mikels-Vigdal A, Garcia CA, Jorgensen B, Velayo AC, Wang R, Adamkewicz JI, Smith V. Selective Allosteric Inhibition of MMP9 Is Efficacious in Preclinical Models of Ulcerative Colitis and Colorectal Cancer. PLoS One 2015; 10:e0127063. [PMID: 25961845 PMCID: PMC4427291 DOI: 10.1371/journal.pone.0127063] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 04/11/2015] [Indexed: 12/20/2022] Open
Abstract
Expression of matrix metalloproteinase 9 (MMP9) is elevated in a variety of inflammatory and oncology indications, including ulcerative colitis and colorectal cancer. MMP9 is a downstream effector and an upstream mediator of pathways involved in growth and inflammation, and has long been viewed as a promising therapeutic target. However, previous efforts to target matrix metalloproteinases (MMPs), including MMP9, have utilized broad-spectrum or semi-selective inhibitors. While some of these drugs showed signs of efficacy in patients, all MMP-targeted inhibitors have been hampered by dose-limiting toxicity or insufficient clinical benefit, likely due to their lack of specificity. Here, we show that selective inhibition of MMP9 did not induce musculoskeletal syndrome (a characteristic toxicity of pan-MMP inhibitors) in a rat model, but did reduce disease severity in a dextran sodium sulfate-induced mouse model of ulcerative colitis. We also found that MMP9 inhibition decreased tumor growth and metastases incidence in a surgical orthotopic xenograft model of colorectal carcinoma, and that inhibition of either tumor- or stroma-derived MMP9 was sufficient to reduce primary tumor growth. Collectively, these data suggest that selective MMP9 inhibition is a promising therapeutic strategy for treatment of inflammatory and oncology indications in which MMP9 is upregulated and is associated with disease pathology, such as ulcerative colitis and colorectal cancer. In addition, we report the development of a potent and highly selective allosteric MMP9 inhibitor, the humanized monoclonal antibody GS-5745, which can be used to evaluate the therapeutic potential of MMP9 inhibition in patients.
Collapse
MESH Headings
- Allosteric Regulation
- Animals
- Antibodies, Monoclonal, Humanized/biosynthesis
- Antibodies, Monoclonal, Humanized/isolation & purification
- Antibodies, Monoclonal, Humanized/pharmacology
- Antineoplastic Agents/isolation & purification
- Antineoplastic Agents/metabolism
- Antineoplastic Agents/pharmacology
- Colitis, Ulcerative/chemically induced
- Colitis, Ulcerative/drug therapy
- Colitis, Ulcerative/enzymology
- Colitis, Ulcerative/genetics
- Colorectal Neoplasms/drug therapy
- Colorectal Neoplasms/enzymology
- Colorectal Neoplasms/genetics
- Colorectal Neoplasms/pathology
- Dextran Sulfate
- Disease Models, Animal
- Drug Evaluation, Preclinical
- Epitope Mapping
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Hybridomas/immunology
- Male
- Matrix Metalloproteinase 9/administration & dosage
- Matrix Metalloproteinase 9/genetics
- Matrix Metalloproteinase 9/metabolism
- Matrix Metalloproteinase Inhibitors/isolation & purification
- Matrix Metalloproteinase Inhibitors/metabolism
- Matrix Metalloproteinase Inhibitors/pharmacology
- Mice
- Mice, Nude
- Rats
- Rats, Inbred Lew
- Recombinant Proteins/administration & dosage
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Signal Transduction
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Derek C. Marshall
- Department of Biology, Gilead Sciences, Inc., Foster City, California, United States of America
| | - Susan K. Lyman
- Department of Biology, Gilead Sciences, Inc., Foster City, California, United States of America
| | - Scott McCauley
- Department of Biology, Gilead Sciences, Inc., Foster City, California, United States of America
| | - Maria Kovalenko
- Department of Biology, Gilead Sciences, Inc., Foster City, California, United States of America
| | - Rhyannon Spangler
- Department of Biology, Gilead Sciences, Inc., Foster City, California, United States of America
| | - Chian Liu
- Department of Biology, Gilead Sciences, Inc., Foster City, California, United States of America
| | - Michael Lee
- Department of Biology, Gilead Sciences, Inc., Foster City, California, United States of America
| | - Christopher O’Sullivan
- Department of Biology, Gilead Sciences, Inc., Foster City, California, United States of America
| | - Vivian Barry-Hamilton
- Department of Biology, Gilead Sciences, Inc., Foster City, California, United States of America
| | - Haben Ghermazien
- Department of Biology, Gilead Sciences, Inc., Foster City, California, United States of America
| | - Amanda Mikels-Vigdal
- Department of Biology, Gilead Sciences, Inc., Foster City, California, United States of America
| | - Carlos A. Garcia
- Department of Process Development, Gilead Sciences, Inc., Oceanside, California, United States of America
| | - Brett Jorgensen
- Department of Biology, Gilead Sciences, Inc., Foster City, California, United States of America
| | - Arleene C. Velayo
- Department of Process Development, Gilead Sciences, Inc., Oceanside, California, United States of America
| | - Ruth Wang
- Department of Biology, Gilead Sciences, Inc., Foster City, California, United States of America
| | - Joanne I. Adamkewicz
- Department of Biology, Gilead Sciences, Inc., Foster City, California, United States of America
| | - Victoria Smith
- Department of Biology, Gilead Sciences, Inc., Foster City, California, United States of America
| |
Collapse
|
49
|
Fabre B, Ramos A, de Pascual-Teresa B. Targeting Matrix Metalloproteinases: Exploring the Dynamics of the S1′ Pocket in the Design of Selective, Small Molecule Inhibitors. J Med Chem 2014; 57:10205-19. [DOI: 10.1021/jm500505f] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Benjamin Fabre
- Departamento de Química
y Bioquímica, Facultad de Farmacia, Universidad CEU San Pablo, Urbanización Monteprincipe, 28668 Madrid, Spain
| | - Ana Ramos
- Departamento de Química
y Bioquímica, Facultad de Farmacia, Universidad CEU San Pablo, Urbanización Monteprincipe, 28668 Madrid, Spain
| | - Beatriz de Pascual-Teresa
- Departamento de Química
y Bioquímica, Facultad de Farmacia, Universidad CEU San Pablo, Urbanización Monteprincipe, 28668 Madrid, Spain
| |
Collapse
|
50
|
Akagi R, Sasho T, Saito M, Endo J, Yamaguchi S, Muramatsu Y, Mukoyama S, Akatsu Y, Katsuragi J, Fukawa T, Takahashi K. Effective knock down of matrix metalloproteinase-13 by an intra-articular injection of small interfering RNA (siRNA) in a murine surgically-induced osteoarthritis model. J Orthop Res 2014; 32:1175-80. [PMID: 24848439 DOI: 10.1002/jor.22654] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 05/02/2014] [Indexed: 02/04/2023]
Abstract
This study investigated the effect of MMP-13 gene knock down on cartilage degradation by injecting small interfering RNA (siRNA) into knee joints in a mouse model of osteoarthritis (OA). OA was induced in male C57BL/6 mice by destabilization of medial meniscus (DMM) surgery. Change of Mmp13 expression over time was determined by qPCR analysis from 3 days to 6 weeks after surgery. Mmp13 and control chemically modified siRNA were injected into the knee joint 1 week after surgery and expression levels were assessed in synovium by qPCR 48 h later. Cartilage degradation was histologically assessed 8 weeks after DMM surgery according to OARSI recommendations. Mmp13 expression levels were elevated 1 week after surgery and peaked at 77 fold at 2 weeks compared to expression at 3 days. A 55% decrease of Mmp13 levels in cartilage was observed 48 h after injection of Mmp13 siRNA (p = 0.05). Significant reduction in the histological score at 8 weeks after surgery was observed in the Mmp13 siRNA-treated group compared to the control siRNA group (p < 0.001). Intra-articular injection of Mmp13 siRNA at the early phase of OA development resulted in effective knock down of Mmp13 expression and delay in cartilage degradation in vivo.
Collapse
Affiliation(s)
- Ryuichiro Akagi
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana Chuo-ku, Chiba, Chiba, 260-8670, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|