1
|
Garg P, Shokrollahi P, Phan CM, Jones L. Biodegradable 3D-Printed Conjunctival Inserts for the Treatment of Dry Eyes. Polymers (Basel) 2025; 17:623. [PMID: 40076115 PMCID: PMC11902855 DOI: 10.3390/polym17050623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
PURPOSE To fabricate 3D-printed, biodegradable conjunctival gelatin methacrylate (GelMA) inserts that can release polyvinyl alcohol (PVA) when exposed to an ocular surface enzyme. METHOD In this work, biodegradable conjunctival inserts were 3D-printed using a stereolithography-based technique. The release of PVA from these insert formulations (containing 10% GelMA and 5% PVA (P-Gel-5%)) was assessed along with different mathematical models of drug release. The biodegradation rates of these inserts were studied in the presence of a tear-film enzyme (matrix metalloproteinase-9; MMP9). The morphology of the inserts before and after enzymatic degradation was monitored using scanning electron microscopy. RESULTS The 3D-printed P-Gel-5% inserts formed a semi-interpenetrating network, which was mechanically stronger than GelMA inserts. The PVA release graphs demonstrate that at the end of 24 h, 222.7 ± 20.3 µg, 265.5 ± 27.1 µg, and 242.7 ± 30.4 µg of PVA were released when exposed to 25, 50, and 100 µg/mL of MMP9, respectively. The release profiles of the P-Gel-5% containing hydrogels in the presence of different concentrations of MMP9 showed the highest linearity with the Korsmeyer-Peppas model. The results suggest that the degradation rate over 24 h is a function of MMP9 enzyme concentration. Over 80% of P-Gel-5% inserts were degraded at the end of 8 h, 12 h, and 24 h in the presence of 100, 50, and 25 µg/mL MMP9 enzyme solutions, respectively. CONCLUSIONS These results demonstrate the potential for 3D printing of GelMA for use as conjunctival inserts. These inserts could be used to deliver PVA, which is a well-known therapeutic agent for dry eye disease. PVA release is influenced by multiple mechanisms, including diffusion and enzymatic degradation, which is supported by morphological studies and biodegradation results.
Collapse
Affiliation(s)
- Piyush Garg
- Centre for Ocular Research & Education (CORE), School of Optometry & Vision Science, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada; (P.G.); (P.S.); (C.-M.P.)
| | - Parvin Shokrollahi
- Centre for Ocular Research & Education (CORE), School of Optometry & Vision Science, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada; (P.G.); (P.S.); (C.-M.P.)
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong
| | - Chau-Minh Phan
- Centre for Ocular Research & Education (CORE), School of Optometry & Vision Science, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada; (P.G.); (P.S.); (C.-M.P.)
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong
| | - Lyndon Jones
- Centre for Ocular Research & Education (CORE), School of Optometry & Vision Science, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada; (P.G.); (P.S.); (C.-M.P.)
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong
| |
Collapse
|
2
|
Fan J, Cheney PP, Bloch S, Xu B, Liang K, Odonkor CA, Edwards WB, Basak S, Mintz R, Biswas P, Achilefu S. Multifunctional Thio-Stabilized Gold Nanoparticles for Near-Infrared Fluorescence Detection and Imaging of Activated Caspase-3. CURR ANAL CHEM 2021; 17:1182-1193. [PMID: 34393690 DOI: 10.2174/1573411017999210112175743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background Gold nanoparticles (AuNPs) are commonly used in nanomedicine because of their unique spectral properties, chemical and biological stability, and ability to quench the fluorescence of organic dyes attached to their surfaces. However, the utility of spherical AuNPs for activatable fluorescence sensing of molecular processes have been confined to resonance-matched fluorophores in the 500 nm to 600 nm spectral range to maximize dye fluorescence quenching efficiency. Expanding the repertoire of fluorophore systems into the NIR fluorescence regimen with emission >800 nm will facilitate the analysis of multiple biological events with high detection sensitivity. Objective The primary goal of this study is to determine if spherical AuNP-induced radiative rate suppression of non-resonant near-infrared (NIR) fluorescent probes can serve as a versatile nanoconstruct for highly sensitive detection and imaging of activated caspase-3 in aqueous media and cancer cells. This required the development of activatable NIR fluorescence sensors of caspase-3 designed to overcome the nonspecific degradation and release of the surface coatings in aqueous media. Method We harnessed the fluorescence-quenching properties and multivalency of spherical AuNPs to develop AuNP-templated activatable NIR fluorescent probes to detect activated caspase-3, an intracellular reporter of early cell death. Freshly AuNPs were coated with a multifunctional NIR fluorescent dye-labeled peptide (LS422) consisting of an RGD peptide sequence that targets αvβ3-integrin protein (αvβ3) on the surface of cancer cells to mediate the uptake and internalization of the sensors in tumor cells; a DEVD peptide sequence for reporting the induction of cell death through caspase-3 mediated NIR fluorescence enhancement; and a multidentate hexacysteine sequence for enhancing self-assembly and stabilizing the multifunctional construct on AuNPs. The integrin binding affinity of LS422 and caspase-3 kinetics were determined by a radioligand competitive binding and fluorogenic peptide assays, respectively. Detection of intracellular caspase-3, cell viability, and the internalization of LS422 in cancer cells were determined by confocal NIR fluorescence spectroscopy and microscopy. Results Narrow size AuNPs (13 nm) were prepared and characterized by transmission electron microscopy and dynamic light scattering. When assembled on the AuNPs, the binding constant of LS422 for αvβ3 improved 11-fold from 13.2 nM to 1.2 nM. Whereas the catalytic turnover of caspase-3 by LS422-AuNPs was similar to the reference fluorogenic peptide, the binding affinity for the enzyme increased by a factor of 2. Unlike the αvβ3 positive, but caspase-3 negative breast cancer MCF-7 cells, treatment of the αvβ3 and caspase-3 positive lung cancer A549 cells with Paclitaxel showed significant fluorescence enhancement within 30 minutes, which correlated with caspase-3 specific activation of LS422-AuNPs fluorescence. Incorporation of a 3.5 mW NIR laser source into our spectrofluorometer increased the detection sensitivity by an order of magnitude (limit of detection ~0.1 nM of cypate) and significantly decreased the signal noise relative to a xenon lamp. This gain in sensitivity enabled the detection of substrate hydrolysis at a broad range of inhibitor concentrations without photobleaching the cypate dye. Conclusion The multifunctional AuNPs demonstrate the use of a non-resonant quenching strategy to design activatable NIR fluorescence molecular probes. The nanoconstruct offers a selective reporting method for detecting activated caspase-3, imaging of cell viability, identifying dying cells, and visualizing the functional status of intracellular enzymes. Performing these tasks with NIR fluorescent probes creates an opportunity to translate the in vitro and cellular analysis of enzymes into in vivo interrogation of their functional status using deep tissue penetrating NIR fluorescence analytical methods.
Collapse
Affiliation(s)
- J Fan
- Departments of Radiology, Washington University School of Medicine, St Louis, United States
| | - P P Cheney
- Departments of Radiology, Washington University School of Medicine, St Louis, United States
| | - S Bloch
- Departments of Radiology, Washington University School of Medicine, St Louis, United States
| | - B Xu
- Departments of Radiology, Washington University School of Medicine, St Louis, United States
| | - K Liang
- Departments of Radiology, Washington University School of Medicine, St Louis, United States
| | - C A Odonkor
- Departments of Radiology, Washington University School of Medicine, St Louis, United States
| | - W B Edwards
- Departments of Radiology, Washington University School of Medicine, St Louis, United States
| | - S Basak
- Department of Energy, Environmental & Chemical Engineering, Washington University, St Louis, United States
| | - R Mintz
- Departments of Radiology, Washington University School of Medicine, St Louis, United States.,Department of Energy, Environmental & Chemical Engineering, Washington University, St Louis, United States.,Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St Louis, United States.,Department of Biomedical Engineering, Washington University, St Louis, United States.,Department of Medicine, Washington University, St Louis, United States
| | - P Biswas
- Department of Energy, Environmental & Chemical Engineering, Washington University, St Louis, United States
| | - S Achilefu
- Departments of Radiology, Washington University School of Medicine, St Louis, United States.,Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St Louis, United States.,Department of Biomedical Engineering, Washington University, St Louis, United States.,Department of Medicine, Washington University, St Louis, United States
| |
Collapse
|
3
|
Sato Y, Asawa K, Huang T, Noiri M, Nakamura N, Ekdahl KN, Nilsson B, Ishihara K, Teramura Y. Induction of Spontaneous Liposome Adsorption by Exogenous Surface Modification with Cell-Penetrating Peptide-Conjugated Lipids. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:9711-9723. [PMID: 34342462 DOI: 10.1021/acs.langmuir.1c01072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The use of amphiphilic molecules such as poly(ethylene glycol)-conjugated phospholipid (PEG-lipid) enables incorporation into liposome surfaces by exogenous addition as a result of the self-assembly with lipids. This technique can be applicable for manipulation of both liposomes and cells. In this study, we aimed to characterize Tat peptide (YGRKKRRQRRR)-conjugated PEG-lipids when used to exogenously surface modify liposomes (size: ca. 100 nm). We earlier reported that cells, which were surface modified with Tat peptides conjugated to PEG-lipids could attach spontaneously to material surfaces without any chemical modification. Here, we synthesized different types of Tat-PEG-lipids by combining PEG of different molecular weights (5 and 40 kDa) with different lipids with three acyl chains (myristoyl, palmitoyl, and stearoyl, respectively) and then studied the spontaneous adsorption of modified liposomes onto a substrate surface induced by the different Tat-PEG-lipids. The amount of adsorbed liposomes strongly depended on the number of incorporated Tat-PEG-lipid moieties: a decrease in both the PEG and the acyl chain lengths led to adsorption of higher amounts of liposomes. Furthermore, when a collagenase-cleavable amino acid sequence was inserted between the Tat sequence and the PEG segment, adsorbed liposomes could be harvested from the substrate by collagenase treatment with no difference in desorption efficiency between the different Tat-PEG-lipids. Thus, Tat-PEG-lipid can be a suitable tool for the manipulation of liposomes and cells.
Collapse
Affiliation(s)
- Yuya Sato
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kenta Asawa
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Tianwei Huang
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Makoto Noiri
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Naoko Nakamura
- Department of Bioscience and Engineering, College of Systems Engineering and Science, Shibaura Institute of Technology, Saitama 337-8570, Japan
| | - Kristina N Ekdahl
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden
- Linnaeus Center of Biomaterials Chemistry, Linnaeus University, SE-391 82 Kalmar, Sweden
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden
| | - Kazuhiko Ishihara
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yuji Teramura
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden
- Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba Central Fifth, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| |
Collapse
|
4
|
Lygeros S, Danielides G, Grafanaki K, Riga M. Matrix metalloproteinases and chronic rhinosinusitis with nasal polyposis. Unravelling a puzzle through a systematic review. Rhinology 2021; 59:245-257. [PMID: 33730750 DOI: 10.4193/rhin20.578] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The expression of metalloproteinases (MMPs) in chronic rhinosinusitis with nasal polyposis (CRSwNP) was reviewed in order to investigate their possible use as therapeutical targets and/or biomarkers. METHODOLOGY The differences between CRSwNP and normal controls or CRS without NP, as well as the effects of various treatments on MMPs, tissue inhibitors of MMPs (TIMPs) and MMP/TIMP ratios were considered as primary outcomes. Additional factors reported to affect MMP expression levels were noted as secondary outcomes. Data regarding inflammatory subtypes, patients’ clinical characteristics, controls, laboratory method(s) and origin of samples were also pooled. Studies on 10 or fewer patients or on specimens other than nasal and serum were excluded. RESULTS Forty-three studies were included. Tissue sample origin, allergic rhinitis, smoking, infection, medication intake and primary or recurrent disease should be considered as confounding factors for MMP levels. MMP-1 and -7 were consistently found to be significantly higher in CRSwNP patients than controls. CRSwNP endotypes with distinctly different inflammation patterns seem to present similar MMP-related remodelling patterns. CONCLUSIONS The existing literature has revealed several population and methodology related confounding factors and remains inconclusive regarding the roles of MMPs in CRSwNP pathophysiology and their possible clinical usefulness as biomarkers and therapeutical targets.
Collapse
Affiliation(s)
- S Lygeros
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Patras, Medical School, Patras, Greece
| | - G Danielides
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Patras, Medical School, Patras, Greece
| | - K Grafanaki
- Department of Biochemistry and Department of Dermatology School of Medicine, University of Patras, Greece
| | - M Riga
- Department of Otorhinolaryngology-Head and Neck Surgery, Dammam Medical Complex, Dammam, Kingdom of Saudi Arabia
| |
Collapse
|
5
|
Jia T, Vaganay E, Carpentier G, Coudert P, Guzman-Gonzales V, Manuel R, Eymin B, Coll JL, Ruggiero F. A collagen Vα1-derived fragment inhibits FGF-2 induced-angiogenesis by modulating endothelial cells plasticity through its heparin-binding site. Matrix Biol 2020; 94:18-30. [DOI: 10.1016/j.matbio.2020.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 01/22/2023]
|
6
|
Sheak JR, Jones DT, Lantz BJ, Maston LD, Vigil D, Resta TC, Resta MM, Howard TA, Kanagy NL, Guo Y, Jankowska-Gan E, Sullivan JA, Braun RK, Burlingham WJ, Gonzalez Bosc LV. NFATc3 regulation of collagen V expression contributes to cellular immunity to collagen type V and hypoxic pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2020; 319:L968-L980. [PMID: 32997513 DOI: 10.1152/ajplung.00184.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic hypoxia (CH)-induced pulmonary hypertension (PH) results, in part, from T helper-17 (TH17) cell-mediated perivascular inflammation. However, the antigen(s) involved is unknown. Cellular immunity to collagen type V (col V) develops after ischemia-reperfusion injury during lung transplant and is mediated by naturally occurring (n)TH17 cells. Col5a1 gene codifies for the α1-helix of col V, which is normally hidden from the immune system within type I collagen in the extracellular matrix. COL5A1 promoter analysis revealed nuclear factor of activated T cells, cytoplasmic 3 (NFATc3) binding sites. Therefore, we hypothesized that smooth muscle NFATc3 upregulates col V expression, leading to nTH17 cell-mediated autoimmunity to col V in response to CH, representing an upstream mechanism in PH development. To test our hypothesis, we measured indexes of PH in inducible smooth muscle cell (SMC)-specific NFATc3 knockout (KO) mice exposed to either CH (380 mmHg) or normoxia and compared them with wild-type (WT) mice. KO mice did not develop PH. In addition, COL5A1 was one of the 1,792 genes differentially affected by both CH and SMC NFATc3 in isolated intrapulmonary arteries, which was confirmed by RT-PCR and immunostaining. Cellular immunity to col V was determined using a trans vivo delayed-type hypersensitivity assay (Tv-DTH). Tv-DTH response was evident only when splenocytes were used from control mice exposed to CH but not from KO mice, and mediated by nTH17 cells. Our results suggest that SMC NFATc3 is important for CH-induced PH in adult mice, in part, by regulating the expression of the lung self-antigen COL5A1 protein contributing to col V-reactive nTH17-mediated inflammation and hypertension.
Collapse
Affiliation(s)
- Joshua R Sheak
- Department of Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - David T Jones
- Department of Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Benjamin J Lantz
- Department of Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Levi D Maston
- Department of Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Danielle Vigil
- Department of Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Thomas C Resta
- Department of Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Micaela M Resta
- Department of Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Tamara A Howard
- Department of Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nancy L Kanagy
- Department of Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Yan Guo
- Department of Internal Medicine, Bioinformatics Shared Resource Center, Division of Molecular Medicine, University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Ewa Jankowska-Gan
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Jeremy A Sullivan
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Rudolf K Braun
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - William J Burlingham
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Laura V Gonzalez Bosc
- Department of Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
7
|
Cerofolini L, Fragai M, Luchinat C. Mechanism and Inhibition of Matrix Metalloproteinases. Curr Med Chem 2019; 26:2609-2633. [PMID: 29589527 DOI: 10.2174/0929867325666180326163523] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/06/2018] [Accepted: 03/06/2018] [Indexed: 01/02/2023]
Abstract
Matrix metalloproteinases hydrolyze proteins and glycoproteins forming the extracellular matrix, cytokines and growth factors released in the extracellular space, and membrane-bound receptors on the outer cell membrane. The pathological relevance of MMPs has prompted the structural and functional characterization of these enzymes and the development of synthetic inhibitors as possible drug candidates. Recent studies have provided a better understanding of the substrate preference of the different members of the family, and structural data on the mechanism by which these enzymes hydrolyze the substrates. Here, we report the recent advancements in the understanding of the mechanism of collagenolysis and elastolysis, and we discuss the perspectives of new therapeutic strategies for targeting MMPs.
Collapse
Affiliation(s)
- Linda Cerofolini
- Magnetic Resonance Center (CERM), University of Florence, and Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Marco Fragai
- Magnetic Resonance Center (CERM), University of Florence, and Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Claudio Luchinat
- Magnetic Resonance Center (CERM), University of Florence, and Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
8
|
Fields GB. Mechanisms of Action of Novel Drugs Targeting Angiogenesis-Promoting Matrix Metalloproteinases. Front Immunol 2019; 10:1278. [PMID: 31214203 PMCID: PMC6558196 DOI: 10.3389/fimmu.2019.01278] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 05/20/2019] [Indexed: 12/16/2022] Open
Abstract
Angiogenesis is facilitated by the proteolytic activities of members of the matrix metalloproteinase (MMP) family. More specifically, MMP-9 and MT1-MMP directly regulate angiogenesis, while several studies indicate a role for MMP-2 as well. The correlation of MMP activity to tumor angiogenesis has instigated numerous drug development programs. However, broad-based and Zn2+-chelating MMP inhibitors have fared poorly in the clinic. Selective MMP inhibition by antibodies, biologicals, and small molecules has utilized unique modes of action, such as (a) binding to protease secondary binding sites (exosites), (b) allosterically blocking the protease active site, or (c) preventing proMMP activation. Clinical trials have been undertaken with several of these inhibitors, while others are in advanced pre-clinical stages. The mechanistically non-traditional MMP inhibitors offer treatment strategies for tumor angiogenesis that avoid the off-target toxicities and lack of specificity that plagued Zn2+-chelating inhibitors.
Collapse
Affiliation(s)
- Gregg B Fields
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, FL, United States.,Department of Chemistry, The Scripps Research Institute/Scripps Florida, Jupiter, FL, United States
| |
Collapse
|
9
|
Hachem RR. The impact of non-HLA antibodies on outcomes after lung transplantation and implications for therapeutic approaches. Hum Immunol 2019; 80:583-587. [PMID: 31005400 DOI: 10.1016/j.humimm.2019.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/09/2019] [Accepted: 04/13/2019] [Indexed: 01/12/2023]
Abstract
The role of donor-specific antibodies (DSA) to mismatched human leukocyte antigens (HLA) in lung allograft rejection has been recognized over the past 20 years. During this time, there has been growing experience and recognition of an important role for non-HLA antibodies in lung allograft rejection. Multiple self-antigens have been identified that elicit autoimmune responses including collagen V, K-α 1 tubulin, angiotensin type 1 receptor, and endothelin type A receptor, but it is likely that other antigens elicit similar responses. The paradigm for the pathogenesis of these autoimmune responses consists of exposure of sequestered self-antigens followed by loss of peripheral tolerance, which then promotes allograft rejection. Studies have focused mainly on the impact of autoimmune responses on the development of Bronchiolitis Obliterans Syndrome or its mouse model surrogate. However, there are emerging data that illustrate that non-HLA antibodies can induce acute antibody-mediated rejection (AMR) after lung transplantation. Treatment has focused on antibody-depletion protocols, but experience is limited to cohort studies and appropriate controlled trials have not been conducted. It is noteworthy that depletion of non-HLA antibodies has been associated with favorable clinical outcomes. Clearly, additional studies are needed to identify the optimal therapeutic approaches to non-HLA antibodies in clinical practice.
Collapse
Affiliation(s)
- Ramsey R Hachem
- Washington University School of Medicine, Division of Pulmonary & Critical Care, 4523 Clayton Ave., Campus Box 8052, St. Louis, MO 63110, United States.
| |
Collapse
|
10
|
Fields GB. Methods for the Construction of Collagen-Based Triple-Helical Peptides Designed as Matrix Metalloproteinase Inhibitors. Methods Mol Biol 2019; 1944:229-252. [PMID: 30840247 DOI: 10.1007/978-1-4939-9095-5_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The triple-helical structure of collagen has been accurately reproduced in numerous chemical and recombinant model systems. Triple-helical peptides have found application for dissecting collagen-stabilizing forces, isolating receptor and protein binding sites in collagen, evaluating collagen-mediated cell signaling activities, mechanistic examination of collagenolytic proteases, and developing novel biomaterials and drug delivery vehicles. Due to their inherent stability to general proteolysis, triple-helical peptides present an opportunity as in vivo inhibitory agents. The present chapter provides methods for the construction of collagen-based triple-helical peptides designed as matrix metalloproteinase inhibitors.
Collapse
Affiliation(s)
- Gregg B Fields
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, FL, USA.
- Department of Chemistry, Scripps Research, Jupiter, FL, USA.
| |
Collapse
|
11
|
Pahwa S, Bhowmick M, Amar S, Cao J, Strongin AY, Fridman R, Weiss SJ, Fields GB. Characterization and regulation of MT1-MMP cell surface-associated activity. Chem Biol Drug Des 2018; 93:1251-1264. [PMID: 30480376 DOI: 10.1111/cbdd.13450] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 10/18/2018] [Accepted: 11/11/2018] [Indexed: 12/19/2022]
Abstract
Quantitative assessment of MT1-MMP cell surface-associated proteolytic activity remains undefined. Presently, MT1-MMP was stably expressed and a cell-based FRET assay developed to quantify activity toward synthetic collagen-model triple-helices. To estimate the importance of cell surface localization and specific structural domains on MT1-MMP proteolysis, activity measurements were performed using a series of membrane-anchored MT1-MMP mutants and compared directly with those of soluble MT1-MMP. MT1-MMP activity (kcat /KM ) on the cell surface was 4.8-fold lower compared with soluble MT1-MMP, with the effect largely manifested in kcat . Deletion of the MT1-MMP cytoplasmic tail enhanced cell surface activity, with both kcat and KM values affected, while deletion of the hemopexin-like domain negatively impacted KM and increased kcat . Overall, cell surface localization of MT1-MMP restricts substrate binding and protein-coupled motions (based on changes in both kcat and KM ) for catalysis. Comparison of soluble and cell surface-bound MT2-MMP revealed 12.9-fold lower activity on the cell surface. The cell-based assay was utilized for small molecule and triple-helical transition state analog MMP inhibitors, which were found to function similarly in solution and at the cell surface. These studies provide the first quantitative assessments of MT1-MMP activity and inhibition in the native cellular environment of the enzyme.
Collapse
Affiliation(s)
- Sonia Pahwa
- Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida
| | - Manishabrata Bhowmick
- Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida
| | - Sabrina Amar
- Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida.,Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, Florida
| | - Jian Cao
- Departments of Medicine/Cancer Prevention and Pathology, Stony Brook University, Stony Brook, New York
| | - Alex Y Strongin
- Cancer Research Center, Sanford Burnham Prebys Medical Research Institute, La Jolla, California
| | - Rafael Fridman
- Department of Pathology and the Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Stephen J Weiss
- Division of Molecular Medicine & Genetics, Department of Internal Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, Michigan
| | - Gregg B Fields
- Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida.,Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, Florida.,The Scripps Research Institute/Scripps Florida, Jupiter, Florida
| |
Collapse
|
12
|
Takai M, Hoy CFO, Yoshihara A. Electrospun Polymeric Microfiber Substrates for Rapid Protein and Cell-based Assays. J PHOTOPOLYM SCI TEC 2018. [DOI: 10.2494/photopolymer.31.65] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
13
|
Determining the Substrate Specificity of Matrix Metalloproteases using Fluorogenic Peptide Substrates. Methods Mol Biol 2018. [PMID: 28299736 DOI: 10.1007/978-1-4939-6863-3_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2023]
Abstract
A continuous assay method, such as the one that utilizes an increase in fluorescence upon hydrolysis, allows for rapid and convenient kinetic evaluation of proteases. To better understand MMP behaviors toward native substrates, a variety of fluorescence resonance energy transfer (FRET)/intramolecular fluorescence energy transfer (IFET) triple-helical substrates have been constructed to examine the collagenolytic activity of MMP family members. Results of these studies have been valuable for providing insights into (a) the relative triple-helical peptidase activities of the various collagenolytic MMPs, (b) the collagen preferences of these MMPs, and (c) the relative roles of MMP domains and specific residues in efficient collagenolysis. The present chapter provides an overview of MMP FRET triple-helical substrates and describes how to construct and utilize these substrates.
Collapse
|
14
|
Yoshihara A, Sekine R, Ueki T, Kondo Y, Sunaga Y, Nakaji-Hirabayashi T, Teramura Y, Takai M. Rapid and highly efficient capture and release of cancer cells using polymeric microfibers immobilized with enzyme-cleavable peptides. Acta Biomater 2018; 67:32-41. [PMID: 29223702 DOI: 10.1016/j.actbio.2017.11.055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 11/23/2017] [Accepted: 11/30/2017] [Indexed: 12/18/2022]
Abstract
Circulating tumor cells (CTCs) are tumor cells present in the blood. CTCs have attracted much attention as a new tumor marker, because their analysis provides useful information for monitoring cancer progress. In this study, we developed cell-capture and release methods using three-dimensional (3D) microfiber fabrics without damaging the cells. Using functional peptides containing sequences from a polystyrene-binding site and a cleavable site for collagenase type IV, immobilized antibodies on the peptides were able to specifically capture MCF-7 cells in a few minutes and release the captured cells from 3D microfiber fabrics incorporating a vacuum system. The efficiency of cell capture was around 80% and that of the cell release was over 90%. The released cells proliferated normally in culture medium, suggesting that our system will be applicable for the culture and analysis of CTCs. STATEMENT OF SIGNIFICANCE In this paper, we report cell-capture and release methods using enzyme-cleavable peptides immobilized on microfiber fabrics which has microporous polymeric three-dimensional structures. Detachment and collection of the selectively captured cancer cells are required for ex vivo culture and their further analysis, whereas the cell detachment methods developed so far might cause cell damage, even if cell viability is high enough. Therefore, specific attachment and gentle detachment from the device are required for the accurate analysis of cells. In this study, for capture and release of cancer cells we designed the peptide cleavable by collagenase type IV, which has no target molecule in cells. Our system will be useful for further CTC analysis and might lead to more accurate cancer diagnosis.
Collapse
|
15
|
Samad AA, Bethry A, Janouskova O, Ciccione J, Wenk C, Coll JL, Subra G, Etrych T, Omar FE, Bakkour Y, Coudane J, Nottelet B. Iterative Photoinduced Chain Functionalization as a Generic Platform for Advanced Polymeric Drug Delivery Systems. Macromol Rapid Commun 2017; 39. [DOI: 10.1002/marc.201700502] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/11/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Assala Al Samad
- IBMM (UMR5247); Univ Montpellier; CNRS; ENSCM; Montpellier France
- Laboratory of applied Chemistry; Faculty of Science III; Lebanese University; P.O. Box 826 Tripoli Lebanon
| | - Audrey Bethry
- IBMM (UMR5247); Univ Montpellier; CNRS; ENSCM; Montpellier France
| | - Olga Janouskova
- Institute of Macromolecular Chemistry; Czech Academy of Sciences; Heyrovského nám. 2 162 06 Prague Czech Republic
| | - Jérémie Ciccione
- IBMM (UMR5247); Univ Montpellier; CNRS; ENSCM; Montpellier France
| | - Christiane Wenk
- INSERM U1209; Institut Albert Bonniot/Université Grenoble Alpes; F-38000 Grenoble France
| | - Jean-Luc Coll
- INSERM U1209; Institut Albert Bonniot/Université Grenoble Alpes; F-38000 Grenoble France
| | - Gilles Subra
- IBMM (UMR5247); Univ Montpellier; CNRS; ENSCM; Montpellier France
| | - Tomas Etrych
- Institute of Macromolecular Chemistry; Czech Academy of Sciences; Heyrovského nám. 2 162 06 Prague Czech Republic
| | - Fawaz El Omar
- Laboratory of applied Chemistry; Faculty of Science III; Lebanese University; P.O. Box 826 Tripoli Lebanon
| | - Youssef Bakkour
- Laboratory of applied Chemistry; Faculty of Science III; Lebanese University; P.O. Box 826 Tripoli Lebanon
| | - Jean Coudane
- IBMM (UMR5247); Univ Montpellier; CNRS; ENSCM; Montpellier France
| | | |
Collapse
|
16
|
Bhowmick M, Tokmina-Roszyk D, Onwuha-Ekpete L, Harmon K, Robichaud T, Fuerst R, Stawikowska R, Steffensen B, Roush W, Wong HR, Fields GB. Second Generation Triple-Helical Peptide Inhibitors of Matrix Metalloproteinases. J Med Chem 2017; 60:3814-3827. [PMID: 28394608 PMCID: PMC6413923 DOI: 10.1021/acs.jmedchem.7b00018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The design of selective matrix metalloproteinase (MMP) inhibitors that also possess favorable solubility properties has proved to be especially challenging. A prior approach using collagen-model templates combined with transition state analogs produced a first generation of triple-helical peptide inhibitors (THPIs) that were effective in vitro against discrete members of the MMP family. These THPI constructs were also highly water-soluble. The present study sought improvements in the first generation THPIs by enhancing thermal stability and selectivity. A THPI selective for MMP-2 and MMP-9 was redesigned to incorporate non-native amino acids (Flp and mep), resulting in an increase of 18 °C in thermal stability. This THPI was effective in vivo in a mouse model of multiple sclerosis, reducing clinical severity and weight loss. Two other THPIs were developed to be more selective within the collagenolytic members of the MMP family. One of these THPIs was serendipitously more effective against MMP-8 than MT1-MMP and was utilized successfully in a mouse model of sepsis. The THPI targeting MMP-8 minimized lung damage, increased production of the anti-inflammatory cytokine IL-10, and vastly improved mouse survival.
Collapse
Affiliation(s)
- Manishabrata Bhowmick
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
- Sigma-Aldrich Corporation, 3 Strathmore Road, Natick, Massachusetts 01760, United States
| | - Dorota Tokmina-Roszyk
- Florida Atlantic University, 5353 Parkside Drive, Jupiter, Florida 33458, United States
| | - Lillian Onwuha-Ekpete
- Florida Atlantic University, 5353 Parkside Drive, Jupiter, Florida 33458, United States
| | - Kelli Harmon
- Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, United States
| | - Trista Robichaud
- University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio Texas 78229, United States
| | - Rita Fuerst
- The Scripps Research Institute/Scripps Florida, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Roma Stawikowska
- Florida Atlantic University, 5353 Parkside Drive, Jupiter, Florida 33458, United States
| | - Bjorn Steffensen
- University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio Texas 78229, United States
- School of Dental Medicine, Tufts University, 1 Kneeland Street, Boston, Massachusetts 02111, United States
| | - William Roush
- The Scripps Research Institute/Scripps Florida, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Hector R. Wong
- Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, 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
| |
Collapse
|
17
|
Matrix Metalloproteinases in Myocardial Infarction and Heart Failure. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 147:75-100. [PMID: 28413032 DOI: 10.1016/bs.pmbts.2017.02.001] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular disease is the leading cause of death, accounting for 600,000 deaths each year in the United States. In addition, heart failure accounts for 37% of health care spending. Matrix metalloproteinases (MMPs) increase after myocardial infarction (MI) and correlate with left ventricular dysfunction in heart failure patients. MMPs regulate the remodeling process by facilitating extracellular matrix turnover and inflammatory signaling. Due to the critical role MMPs play during cardiac remodeling, there is a need to better understand the pathophysiological mechanism of MMPs, including the biological function of the downstream products of MMP proteolysis. Future studies developing new therapeutic targets that inhibit specific MMP actions to limit the development of heart failure post-MI are warranted. This chapter focuses on the role of MMPs post-MI, the efficiency of MMPs as biomarkers for MI or heart failure, and the future of MMPs and their cleavage products as targets for prevention of post-MI heart failure.
Collapse
|
18
|
Early matrix metalloproteinase-9 inhibition post-myocardial infarction worsens cardiac dysfunction by delaying inflammation resolution. J Mol Cell Cardiol 2016; 100:109-117. [PMID: 27746126 DOI: 10.1016/j.yjmcc.2016.10.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 10/10/2016] [Accepted: 10/12/2016] [Indexed: 01/06/2023]
Abstract
Matrix metalloproteinase-9 (MMP-9) is robustly elevated in the first week post-myocardial infarction (MI). Targeted deletion of the MMP-9 gene attenuates cardiac remodeling post-MI by reducing macrophage infiltration and collagen accumulation through increased apoptosis and reduced inflammation. In this study, we used a translational experimental design to determine whether selective MMP-9 inhibition early post-MI would be an effective therapeutic strategy in mice. We enrolled male C57BL/6J mice (3-6months old, n=116) for this study. Mice were subjected to coronary artery ligation. Saline or MMP-9 inhibitor (MMP-9i; 0.03μg/day) treatment was initiated at 3h post-MI and the mice were sacrificed at day (D) 1 or 7 post-MI. MMP-9i reduced MMP-9 activity by 31±1% at D1 post-MI (p<0.05 vs saline) and did not affect survival or infarct area. Surprisingly, MMP-9i treatment increased infarct wall thinning and worsened cardiac function at D7 post-MI. While MMP-9i enhanced neutrophil infiltration at D1 and macrophage infiltration at D7 post-MI, CD36 levels were lower in MMP-9i compared to saline, signifying reduced phagocytic potential per macrophage. Escalation and prolongation of the inflammatory response at D7 post-MI in the MMP-9i group was evident by increased expression of 18 pro-inflammatory cytokines (all p<0.05). MMP-9i reduced cleaved caspase 3 levels at D7 post-MI, consistent with reduced apoptosis and defective inflammation resolution. Because MMP-9i effects on inflammatory cells were significantly different from previously observed MMP-9 null mechanisms, we evaluated pre-MI (baseline) systemic differences between C57BL/6J and MMP-9 null plasma. By mass spectrometry, 34 plasma proteins were significantly different between groups, revealing a previously unappreciated altered baseline environment pre-MI when MMP-9 was deleted. In conclusion, early MMP-9 inhibition delayed inflammation resolution and exacerbated cardiac dysfunction, highlighting the importance of using translational approaches in mice.
Collapse
|
19
|
Knapinska AM, Tokmina-Roszyk D, Amar S, Tokmina-Roszyk M, Mochalin VN, Gogotsi Y, Cosme P, Terentis AC, Fields GB. Solid-phase synthesis, characterization, and cellular activities of collagen-model nanodiamond-peptide conjugates. Biopolymers 2016; 104:186-95. [PMID: 25753561 DOI: 10.1002/bip.22636] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 02/24/2015] [Accepted: 02/24/2015] [Indexed: 01/10/2023]
Abstract
Nanodiamonds (NDs) have received considerable attention as potential drug delivery vehicles. NDs are small (∼5 nm diameter), can be surface modified in a controllable fashion with a variety of functional groups, and have little observed toxicity in vitro and in vivo. However, most biomedical applications of NDs utilize surface adsorption of biomolecules, as opposed to covalent attachment. Covalent modification provides reliable and reproducible ND-biomolecule ratios, and alleviates concerns over biomolecule desorption prior to delivery. The present study has outlined methods for the efficient solid-phase conjugation of ND to peptides and characterization of ND-peptide conjugates. Utilizing collagen-derived peptides, the ND was found to support or even enhance the cell adhesion and viability activities of the conjugated sequence. Thus, NDs can be incorporated into peptides and proteins in a selective manner, where the presence of the ND could potentially enhance the in vivo activities of the biomolecule it is attached to.
Collapse
Affiliation(s)
- Anna M Knapinska
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, FL, 33458.,Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, FL, 34987
| | - Dorota Tokmina-Roszyk
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, FL, 33458.,Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, FL, 34987
| | - Sabrina Amar
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, FL, 33458.,Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, FL, 34987
| | - Michal Tokmina-Roszyk
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, FL, 33458.,Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, FL, 34987
| | - Vadym N Mochalin
- Department of Materials Science and Engineering and A.J. Drexel Nanotechnology Institute, Drexel University, Philadelphia, PA, 19104
| | - Yury Gogotsi
- Department of Materials Science and Engineering and A.J. Drexel Nanotechnology Institute, Drexel University, Philadelphia, PA, 19104
| | - Patrick Cosme
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, FL, 33458
| | - Andrew C Terentis
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, FL, 33458
| | - Gregg B Fields
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, FL, 33458.,Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, FL, 34987.,Department of Chemistry, The Scripps Research Institute/Scripps Florida, Jupiter, FL, 33458
| |
Collapse
|
20
|
Prior SH, Byrne TS, Tokmina-Roszyk D, Fields GB, Van Doren SR. Path to Collagenolysis: COLLAGEN V TRIPLE-HELIX MODEL BOUND PRODUCTIVELY AND IN ENCOUNTERS BY MATRIX METALLOPROTEINASE-12. J Biol Chem 2016; 291:7888-901. [PMID: 26887942 DOI: 10.1074/jbc.m115.703124] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Indexed: 11/06/2022] Open
Abstract
Collagenolysis is essential in extracellular matrix homeostasis, but its structural basis has long been shrouded in mystery. We have developed a novel docking strategy guided by paramagnetic NMR that positions a triple-helical collagen V mimic (synthesized with nitroxide spin labels) in the active site of the catalytic domain of matrix metalloproteinase-12 (MMP-12 or macrophage metalloelastase) primed for catalysis. The collagenolytically productive complex forms by utilizing seven distinct subsites that traverse the entire length of the active site. These subsites bury ∼1,080 Å(2)of surface area, over half of which is contributed by the trailing strand of the synthetic collagen V mimic, which also appears to ligate the catalytic zinc through the glycine carbonyl oxygen of its scissile G∼VV triplet. Notably, the middle strand also occupies the full length of the active site where it contributes extensive interfacial contacts with five subsites. This work identifies, for the first time, the productive and specific interactions of a collagen triple helix with an MMP catalytic site. The results uniquely demonstrate that the active site of the MMPs is wide enough to accommodate two strands from collagen triple helices. Paramagnetic relaxation enhancements also reveal an extensive array of encounter complexes that form over a large part of the catalytic domain. These transient complexes could possibly facilitate the formation of collagenolytically active complexes via directional Brownian tumbling.
Collapse
Affiliation(s)
- Stephen H Prior
- From the Department of Biochemistry, University of Missouri, Columbia, Missouri 65211
| | - Todd S Byrne
- From the Department of Biochemistry, University of Missouri, Columbia, Missouri 65211
| | - Dorota Tokmina-Roszyk
- the Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, Florida 33458, and
| | - Gregg B Fields
- the Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, Florida 33458, and the Scripps Research Institute/Scripps Florida, Jupiter, Florida 33458
| | - Steven R Van Doren
- From the Department of Biochemistry, University of Missouri, Columbia, Missouri 65211,
| |
Collapse
|
21
|
Oudart JB, Brassart-Pasco S, Vautrin A, Sellier C, Machado C, Dupont-Deshorgue A, Brassart B, Baud S, Dauchez M, Monboisse JC, Harakat D, Maquart FX, Ramont L. Plasmin releases the anti-tumor peptide from the NC1 domain of collagen XIX. Oncotarget 2016; 6:3656-68. [PMID: 25668817 PMCID: PMC4414144 DOI: 10.18632/oncotarget.2849] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 12/05/2014] [Indexed: 02/07/2023] Open
Abstract
During tumor invasion, tumor cells degrade the extracellular matrix. Basement membrane degradation is responsible for the production of peptides with anti-tumor properties. Type XIX collagen is associated with basement membranes in vascular, neuronal, mesenchymal and epithelial tissues. Previously, we demonstrated that the non-collagenous NC1, C-terminal, domain of collagen XIX [NC1(XIX)] inhibits the migration capacities of tumor cells and exerts a strong inhibition of tumor growth. Here, we demonstrate that plasmin, one of the most important enzyme involved in tumor invasion, was able to release a fragment of NC1(XIX), which retained the anti-tumor activity. Molecular modeling studies showed that NC1(XIX) and the anti-tumor fragment released by plasmin (F4) adopted locally the same type I β-turn conformation. This suggests that the anti-tumor effect is conformation-dependent. This study demonstrates that collagen XIX is a novel proteolytic substrate for plasmin. Such release may constitute a defense of the organism against tumor invasion.
Collapse
Affiliation(s)
- Jean-Baptiste Oudart
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,CHU de Reims, Laboratoire Central de Biochimie, Reims, France
| | - Sylvie Brassart-Pasco
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Alexia Vautrin
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Christèle Sellier
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Carine Machado
- Université de Reims Champagne-Ardenne, Institut de Chimie Moléculaire de Reims, CNRS UMR N°7312, Faculté de Pharmacie, Reims, France
| | - Aurelie Dupont-Deshorgue
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Bertrand Brassart
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - S Baud
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,Plateau de Modélisation Moléculaire Multi-échelle, UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
| | - Manuel Dauchez
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,Plateau de Modélisation Moléculaire Multi-échelle, UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
| | - Jean-Claude Monboisse
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,CHU de Reims, Laboratoire Central de Biochimie, Reims, France
| | - Dominique Harakat
- Université de Reims Champagne-Ardenne, Institut de Chimie Moléculaire de Reims, CNRS UMR N°7312, Faculté de Pharmacie, Reims, France
| | - François-Xavier Maquart
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,CHU de Reims, Laboratoire Central de Biochimie, Reims, France
| | - Laurent Ramont
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,CHU de Reims, Laboratoire Central de Biochimie, Reims, France
| |
Collapse
|
22
|
Fields GB, Stawikowski MJ. Imaging Matrix Metalloproteinase Activity Implicated in Breast Cancer Progression. Methods Mol Biol 2016; 1406:303-29. [PMID: 26820965 DOI: 10.1007/978-1-4939-3444-7_25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Proteolysis has been cited as an important contributor to cancer initiation and progression. One can take advantage of tumor-associated proteases to selectively deliver imaging agents. Protease-activated imaging systems have been developed using substrates designed for hydrolysis by members of the matrix metalloproteinase (MMP) family. We presently describe approaches by which one can optically image matrix metalloproteinase activity implicated in breast cancer progression, with consideration of selective versus broad protease probes.
Collapse
Affiliation(s)
- Gregg B Fields
- Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, FL, 33458, USA. .,Department of Chemistry, The Scripps Research Institute/Scripps Florida, Jupiter, FL, 33458, USA. .,Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, FL, 34987, USA.
| | - Maciej J Stawikowski
- Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, FL, 33458, USA
| |
Collapse
|
23
|
Stawikowski MJ, Stawikowska R, Fields GB. Collagenolytic Matrix Metalloproteinase Activities toward Peptomeric Triple-Helical Substrates. Biochemistry 2015; 54:3110-21. [PMID: 25897652 DOI: 10.1021/acs.biochem.5b00110] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Although collagenolytic matrix metalloproteinases (MMPs) possess common domain organizations, there are subtle differences in their processing of collagenous triple-helical substrates. In this study, we have incorporated peptoid residues into collagen model triple-helical peptides and examined MMP activities toward these peptomeric chimeras. Several different peptoid residues were incorporated into triple-helical substrates at subsites P3, P1, P1', and P10' individually or in combination, and the effects of the peptoid residues were evaluated on the activities of full-length MMP-1, MMP-8, MMP-13, and MMP-14/MT1-MMP. Most peptomers showed little discrimination between MMPs. However, a peptomer containing N-methyl Gly (sarcosine) in the P1' subsite and N-isobutyl Gly (NLeu) in the P10' subsite was hydrolyzed efficiently only by MMP-13 [nomenclature relative to the α1(I)772-786 sequence]. Cleavage site analysis showed hydrolysis at the Gly-Gln bond, indicating a shifted binding of the triple helix compared to the parent sequence. Favorable hydrolysis by MMP-13 was not due to sequence specificity or instability of the substrate triple helix but rather was based on the specific interactions of the P7' peptoid residue with the MMP-13 hemopexin-like domain. A fluorescence resonance energy transfer triple-helical peptomer was constructed and found to be readily processed by MMP-13, not cleaved by MMP-1 and MMP-8, and weakly hydrolyzed by MT1-MMP. The influence of the triple-helical structure containing peptoid residues on the interaction between MMP subsites and individual substrate residues may provide additional information about the mechanism of collagenolysis, the understanding of collagen specificity, and the design of selective MMP probes.
Collapse
Affiliation(s)
- Maciej J Stawikowski
- †Florida Atlantic University, 5353 Parkside Drive, Jupiter, Florida 33458, United States.,‡Torrey Pines Institute for Molecular Studies, 11350 Southwest Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Roma Stawikowska
- †Florida Atlantic University, 5353 Parkside Drive, Jupiter, Florida 33458, United States.,‡Torrey Pines Institute for Molecular Studies, 11350 Southwest Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Gregg B Fields
- †Florida Atlantic University, 5353 Parkside Drive, Jupiter, Florida 33458, United States.,‡Torrey Pines Institute for Molecular Studies, 11350 Southwest Village Parkway, Port St. Lucie, Florida 34987, United States.,§The Scripps Research Institute/Scripps Florida, 130 Scripps Way, Jupiter, Florida 33458, United States
| |
Collapse
|
24
|
Chow DWY, Chau Y, Yeung WK, Westermeyer HD. In vitroevaluation of the inhibitory effect of canine serum, canine fresh frozen plasma, freeze-thaw-cycled plasma, and Solcoseryl™on matrix metalloproteinases 2 and 9. Vet Ophthalmol 2014; 18:229-33. [DOI: 10.1111/vop.12204] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Derek W. Y. Chow
- Peace Avenue Veterinary Clinic; G/F 7B Liberty Road Homantin Kowloon Hong Kong
| | - Ying Chau
- Division of Biomedical Engineering; Department of Chemical and Biomolecular Engineering; The Hong Kong University of Science and Technology; Clear Water Bay Hong Kong
| | - Wai Kit Yeung
- Bioengineering Graduate Program; Division of Biomedical Engineering; The Hong Kong University of Science and Technology; Clear Water Bay Hong Kong
| | - Hans D. Westermeyer
- Peace Avenue Veterinary Clinic; G/F 7B Liberty Road Homantin Kowloon Hong Kong
| |
Collapse
|
25
|
Zhang X, Bresee J, Fields GB, Edwards WB. Near-infrared triple-helical peptide with quenched fluorophores for optical imaging of MMP-2 and MMP-9 proteolytic activity in vivo. Bioorg Med Chem Lett 2014; 24:3786-3790. [PMID: 25047578 DOI: 10.1016/j.bmcl.2014.06.072] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 06/20/2014] [Accepted: 06/23/2014] [Indexed: 12/15/2022]
Abstract
The gelatinase members of the MMP family have consistently been associated with tumor invasiveness, which make them an attractive target for molecular imaging. We report new activatable proteolytic optical imaging agents that consist of triple-helical peptide (THP) conjugates, with high specificity to the gelatinases, bearing quenched cypate dyes. With quenching efficiencies up to 51%, the amplified fluorescence signal upon cypate3-THP hydrolysis by the gelatinases (kcat/KM values of 6.4×10(3) M(-1) s(-1) to 9.1×10(3) M(-1) s(-1) for MMP-2 and MMP-9, respectively) in mice bearing human fibrosarcoma xenografted tumors was monitored with fluorescence molecular tomography. There was significant fluorescence enhancement within the tumor and this enhancement was reduced by treatment with pan-MMP inhibitor, Ilomastat. These data, combined with the gelatinase substrate specificity observed in vitro, indicated the observed fluorescence at the site of the tumor was due to gelatinase mediated hydrolysis of cypate3-THP.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Jamee Bresee
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Gregg B Fields
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, FL, 34987, USA
| | - W Barry Edwards
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| |
Collapse
|
26
|
Zhang X, Bresee J, Cheney PP, Xu B, Bhowmick M, Cudic M, Fields GB, Edwards WB. Evaluation of a triple-helical peptide with quenched FluorSophores for optical imaging of MMP-2 and MMP-9 proteolytic activity. Molecules 2014; 19:8571-88. [PMID: 24959683 PMCID: PMC4347883 DOI: 10.3390/molecules19068571] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 06/05/2014] [Accepted: 06/11/2014] [Indexed: 01/15/2023] Open
Abstract
Matrix metalloproteinases (MMP) 2 and 9, the gelatinases, have consistently been associated with tumor progression. The development of gelatinase-specific probes will be critical for identifying in vivo gelatinoic activity to understand the molecular role of the gelatinases in tumor development. Recently, a self-assembling homotrimeric triple-helical peptide (THP), incorporating a sequence from type V collagen, with high substrate specificity to the gelatinases has been developed. To determine whether this THP would be suitable for imaging protease activity, 5-carboxyfluorescein (5FAM) was conjugated, resulting in 5FAM3-THP and 5FAM6-THP, which were quenched up to 50%. 5FAM6-THP hydrolysis by MMP-2 and MMP-9 displayed kcat/KM values of 1.5 × 104 and 5.4 × 103 M−1 s−1, respectively. Additionally 5FAM6-THP visualized gelatinase activity in gelatinase positive HT-1080 cells, but not in gelatinase negative MCF-7 cells. Furthermore, the fluorescence in the HT-1080 cells was greatly attenuated by the addition of a MMP-2 and MMP-9 inhibitor, SB-3CT, indicating that the observed fluorescence release was mediated by gelatinase proteolysis and not non-specific proteolysis of the THPs. These results demonstrate that THPs fully substituted with fluorophores maintain their substrate specificity to the gelatinases in human cancer cells and may be useful in in vivo molecular imaging of gelatinase activity.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| | - Jamee Bresee
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| | - Philip P Cheney
- Department of Chemistry and Biochemistry, University of Denver, Denver, CO 80208, USA.
| | - Baogang Xu
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA.
| | - Manishabrata Bhowmick
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA.
| | - Mare Cudic
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA.
| | - Gregg B Fields
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA.
| | | |
Collapse
|
27
|
Tokmina-Roszyk M, Tokmina-Roszyk D, Bhowmick M, Fields GB. Development of a Förster resonance energy transfer assay for monitoring bacterial collagenase triple-helical peptidase activity. Anal Biochem 2014; 453:61-9. [PMID: 24608089 DOI: 10.1016/j.ab.2014.02.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 01/30/2014] [Accepted: 02/23/2014] [Indexed: 11/25/2022]
Abstract
Due to their efficiency in the hydrolysis of the collagen triple helix, Clostridium histolyticum collagenases are used for isolation of cells from various tissues, including isolation of the human pancreatic islets. However, the instability of clostridial collagenase I (Col G) results in a degraded Col G that has weak collagenolytic activity and an adverse effect on islet isolation and viability. A Förster resonance energy transfer triple-helical peptide substrate (fTHP) has been developed for selective evaluation of bacterial collagenase activity. The fTHP [sequence: Gly-mep-Flp-(Gly-Pro-Hyp)4-Gly-Lys(Mca)-Thr-Gly-Pro-Leu-Gly-Pro-Pro-Gly-Lys(Dnp)-Ser-(Gly-Pro-Hyp)4-NH2] had a melting temperature (Tm) of 36.2°C and was hydrolyzed efficiently by bacterial collagenase (k(cat)/K(M)=25,000s(-1)M(-1)) but not by clostripain, trypsin, neutral protease, thermolysin, or elastase. The fTHP bacterial collagenase assay allows for rapid and specific assessment of enzyme activity toward triple helices and, thus, potential application for evaluating the efficiency of cell isolation by collagenases.
Collapse
Affiliation(s)
| | | | | | - Gregg B Fields
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, FL 34987, USA.
| |
Collapse
|
28
|
Lauer JL, Bhowmick M, Tokmina-Roszyk D, Lin Y, Van Doren SR, Fields GB. The role of collagen charge clusters in the modulation of matrix metalloproteinase activity. J Biol Chem 2014; 289:1981-92. [PMID: 24297171 PMCID: PMC3900948 DOI: 10.1074/jbc.m113.513408] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 11/28/2013] [Indexed: 12/22/2022] Open
Abstract
Members of the matrix metalloproteinase (MMP) family selectively cleave collagens in vivo. Several substrate structural features that direct MMP collagenolysis have been identified. The present study evaluated the role of charged residue clusters in the regulation of MMP collagenolysis. A series of 10 triple-helical peptide (THP) substrates were constructed in which either Lys-Gly-Asp or Gly-Asp-Lys motifs replaced Gly-Pro-Hyp (where Hyp is 4-hydroxy-L-proline) repeats. The stabilities of THPs containing the two different motifs were analyzed, and kinetic parameters for substrate hydrolysis by six MMPs were determined. A general trend for virtually all enzymes was that, as Gly-Asp-Lys motifs were moved from the extreme N and C termini to the interior next to the cleavage site sequence, kcat/Km values increased. Additionally, all Gly-Asp-Lys THPs were as good or better substrates than the parent THP in which Gly-Asp-Lys was not present. In turn, the Lys-Gly-Asp THPs were also always better substrates than the parent THP, but the magnitude of the difference was considerably less compared with the Gly-Asp-Lys series. Of the MMPs tested, MMP-2 and MMP-9 most greatly favored the presence of charged residues with preference for the Gly-Asp-Lys series. Lys-Gly-(Asp/Glu) motifs are more commonly found near potential MMP cleavage sites than Gly-(Asp/Glu)-Lys motifs. As Lys-Gly-Asp is not as favored by MMPs as Gly-Asp-Lys, the Lys-Gly-Asp motif appears advantageous over the Gly-Asp-Lys motif by preventing unwanted MMP hydrolysis. More specifically, the lack of Gly-Asp-Lys clusters may diminish potential MMP-2 and MMP-9 collagenolytic activity. The present study indicates that MMPs have interactions spanning the P23-P23' subsites of collagenous substrates.
Collapse
Affiliation(s)
- Janelle L. Lauer
- From the Department of Biochemistry, University of Texas Health Science Center, San Antonio, Texas 78229
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Manishabrata Bhowmick
- From the Department of Biochemistry, University of Texas Health Science Center, San Antonio, Texas 78229
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida 34987 and
| | - Dorota Tokmina-Roszyk
- From the Department of Biochemistry, University of Texas Health Science Center, San Antonio, Texas 78229
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida 34987 and
| | - Yan Lin
- From the Department of Biochemistry, University of Texas Health Science Center, San Antonio, Texas 78229
| | - Steven R. Van Doren
- Department of Biochemistry, University of Missouri, Columbia, Missouri 65211
| | - Gregg B. Fields
- From the Department of Biochemistry, University of Texas Health Science Center, San Antonio, Texas 78229
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida 34987 and
| |
Collapse
|
29
|
Nakaji-Hirabayashi T, Kitano H. Interleukin-10 chimeric protein to protect transplanted neural progenitor cells from immune responses. J Mater Chem B 2014; 2:8598-8607. [DOI: 10.1039/c4tb01413h] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Novel system for protecting transplanted cells from inflammatory responses.
Collapse
Affiliation(s)
| | - H. Kitano
- Department of Applied Chemistry
- Graduate School of Science and Engineering
- University of Toyama
- Toyama 930-8555, Japan
| |
Collapse
|
30
|
Díaz N, Suárez D, Valdés H. Unraveling the molecular structure of the catalytic domain of matrix metalloproteinase-2 in complex with a triple-helical peptide by means of molecular dynamics simulations. Biochemistry 2013; 52:8556-69. [PMID: 24164447 DOI: 10.1021/bi401144p] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Herein, we present the results of a computational study that employed various simulation methodologies to build and validate a series of molecular models of a synthetic triple-helical peptide (fTHP-5) both in its native state and in a prereactive complex with the catalytic domain of the MMP-2 enzyme. First, the structure and dynamical properties of the fTHP-5 substrate are investigated by means of molecular dynamics (MD) simulations. Then, the propensity of each of the three peptide chains in fTHP-5 to be distorted around the scissile peptide bond is assessed by carrying out potential of mean force calculations. Subsequently, the distorted geometries of fTHP-5 are docked within the MMP-2 active site following a semirigid protocol, and the most stable docked structures are fully relaxed and characterized by extensive MD simulations in explicit solvent. Following a similar approach, we also investigate a hypothetical ternary complex formed between two MMP-2 catalytic units and a single fTHP-5 molecule. Overall, our models for the MMP-2/fTHP-5 complexes unveil the extent to which the triple helix is distorted to allow the accommodation of an individual peptide chain within the MMP active site.
Collapse
Affiliation(s)
- Natalia Díaz
- Departamento de Química Física y Analítica, Universidad de Oviedo , Julián Clavería 8, Oviedo (Asturias) 33006, Spain
| | | | | |
Collapse
|
31
|
Hawkins KE, DeMars KM, Yang C, Rosenberg GA, Candelario-Jalil E. Fluorometric immunocapture assay for the specific measurement of matrix metalloproteinase-9 activity in biological samples: application to brain and plasma from rats with ischemic stroke. Mol Brain 2013; 6:14. [PMID: 23522154 PMCID: PMC3620676 DOI: 10.1186/1756-6606-6-14] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 03/19/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Matrix metalloproteinases are important factors in the molecular mechanisms leading to neuronal injury in many neurological disorders. Matrix metalloproteinase (MMP)-9 is up-regulated after cerebral ischemia and neuroinflammation and is actively involved in blood-brain barrier disruption. Current methods of measuring MMP-9 activity, such as gelatin-substrate zymography, are unspecific and arduous. Here we developed an immunocapture assay with high efficiency, specificity, and sensitivity for quantifying endogenously active as well as total MMP-9 activity. RESULTS A fluorescence resonance energy transfer (FRET) peptide-based immunocapture assay was developed that enables the accurate assessment of total and active forms of MMP-9 in complex biological samples. The FRET assay demonstrated correct and efficient binding of MMP-9 to a mouse monoclonal MMP-9 antibody and high specificity of the immunocapture antibody for MMP-9. Total and active levels of MMP-9 were measured in rat brain homogenates, plasma, human HT-1080 conditioned media, and RBE4 endothelial cell lysates. The FRET immunocapture assay yielded highly similar results for total MMP-9 activity when compared to gelatin-substrate zymography. CONCLUSIONS We suggest that the new FRET peptide-based immunocapture assay is a viable replacement of zymography for sensitive and high throughput quantification of MMP-9 activity in biological samples.
Collapse
Affiliation(s)
- Kimberly E Hawkins
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Kelly M DeMars
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Gary A Rosenberg
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
32
|
Knapinska A, Fields GB. Chemical biology for understanding matrix metalloproteinase function. Chembiochem 2012; 13:2002-20. [PMID: 22933318 PMCID: PMC3951272 DOI: 10.1002/cbic.201200298] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Indexed: 12/20/2022]
Abstract
The matrix metalloproteinase (MMP) family has long been associated with normal physiological processes such as embryonic implantation, tissue remodeling, organ development, and wound healing, as well as multiple aspects of cancer initiation and progression, osteoarthritis, inflammatory and vascular diseases, and neurodegenerative diseases. The development of chemically designed MMP probes has advanced our understanding of the roles of MMPs in disease in addition to shedding considerable light on the mechanisms of MMP action. The first generation of protease-activated agents has demonstrated proof of principle as well as providing impetus for in vivo applications. One common problem has been a lack of agent stability at nontargeted tissues and organs due to activation by multiple proteases. The present review considers how chemical biology has impacted the progress made in understanding the roles of MMPs in disease and the basic mechanisms of MMP action.
Collapse
Affiliation(s)
| | - Gregg B. Fields
- Departments of Chemistry and Biology Torrey Pines Institute for Molecular Studies 11350 SW Village Parkway, Port St. Lucie, FL 34987 (USA)
| |
Collapse
|
33
|
|
34
|
Nahire R, Paul S, Scott MD, Singh RK, Muhonen WW, Shabb J, Gange KN, Srivastava DK, Sarkar K, Mallik S. Ultrasound enhanced matrix metalloproteinase-9 triggered release of contents from echogenic liposomes. Mol Pharm 2012; 9:2554-64. [PMID: 22849291 DOI: 10.1021/mp300165s] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The extracellular enzyme matrix metalloproteinase-9 (MMP-9) is overexpressed in atherosclerotic plaques and in metastatic cancers. The enzyme is responsible for rupture of the plaques and for the invasion and metastasis of a large number of cancers. The ability of ultrasonic excitation to induce thermal and mechanical effects has been used to release drugs from different carriers. However, the majority of these studies were performed with low frequency ultrasound (LFUS) at kilohertz frequencies. Clinical usage of LFUS excitations will be limited due to harmful biological effects. Herein, we report our results on the release of encapsulated contents from substrate lipopeptide incorporated echogenic liposomes triggered by recombinant human MMP-9. The contents release was further enhanced by the application of diagnostic frequency (3 MHz) ultrasound. The echogenic liposomes were successfully imaged employing a medical ultrasound transducer (4-15 MHz). The conditioned cell culture media from cancer cells (secreting MMP-9) released the encapsulated dye from the liposomes (30-50%), and this release is also increased (50-80%) by applying diagnostic frequency ultrasound (3 MHz) for 3 min. With further developments, these liposomes have the potential to serve as multimodal carriers for triggered release and simultaneous ultrasound imaging.
Collapse
Affiliation(s)
- Rahul Nahire
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Akers WJ, Xu B, Lee H, Sudlow GP, Fields GB, Achilefu S, Edwards WB. Detection of MMP-2 and MMP-9 activity in vivo with a triple-helical peptide optical probe. Bioconjug Chem 2012; 23:656-63. [PMID: 22309692 DOI: 10.1021/bc300027y] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We report a novel activatable NIR fluorescent probe for in vivo detection of cancer-related matrix metalloproteinase (MMP) activity. The probe is based on a triple-helical peptide substrate (THP) with high specificity for MMP-2 and MMP-9 relative to other members of the MMP family. MMP-2 and MMP-9 (also known as gelatinases) are specifically associated with cancer cell invasion and cancer-related angiogenesis. At the center of each 5 kDa peptide strand is a gelatinase sensitive sequence flanked by 2 Lys residues conjugated with NIR fluorescent dyes. Upon self-assembly of the triple-helical structure, the 3 peptide chains intertwine, bringing the fluorophores into close proximity and reducing fluorescence via quenching. Upon enzymatic cleavage of the triple-helical peptide, 6 labeled peptide chains are released, resulting in an amplified fluorescent signal. The fluorescence yield of the probe increases 3.8-fold upon activation. Kinetic analysis showed a rate of LS276-THP hydrolysis by MMP-2 (k(cat)/K(M) = 30,000 s(-1) M(-1)) similar to that of MMP-2 catalysis of an analogous fluorogenic THP. Administration of LS276-THP to mice bearing a human fibrosarcoma xenografted tumor resulted in a tumor fluorescence signal more than 5-fold greater than that of muscle. This signal enhancement was reduced by treatment with the MMP inhibitor Ilomostat, indicating that the observed tumor fluorescence was indeed enzyme mediated. These results are the first to demonstrate that triple-helical peptides are suitable for highly specific in vivo detection of tumor-related MMP-2 and MMP-9 activity.
Collapse
Affiliation(s)
- Walter J Akers
- Mallinckrodt Institute of Radiology, Washington University, School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Tiriveedhi V, Angaswamy N, Brand D, Weber J, Gelman AG, Hachem R, Trulock EP, Meyers B, Patterson G, Mohanakumar T. A shift in the collagen V antigenic epitope leads to T helper phenotype switch and immune response to self-antigen leading to chronic lung allograft rejection. Clin Exp Immunol 2012; 167:158-68. [PMID: 22132895 DOI: 10.1111/j.1365-2249.2011.04486.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Immune responses to human leucocyte antigen (HLA) and self-antigen collagen V (Col-V) have been proposed in the pathogenesis of chronic rejection (bronchiolitis obliterans syndrome, BOS) following human lung transplantation (LTx). In this study, we defined the role for the shift in immunodominant epitopes of Col-V in inducing T helper phenotype switch leading to immunity to Col-V and BOS. Sera and lavage from BOS(+) LTx recipients with antibodies to Col-V were analysed. Two years prior to BOS, patients developed antibodies to both Col-V,α1(V) and α2(V) chains. However, at clinical diagnosis of BOS, antibodies became restricted to α1(V). Further, lung biopsy from BOS(+) patients bound to antibodies to α1(V), indicating that these epitopes are exposed. Fourteen Col-V peptides [pep1-14, pep1-4 specific to α1(V), pep5-8 to α1,2(V) and pep9-14 to α2(V)] which bind to HLA-DR4 and -DR7, demonstrated that prior to BOS, pep 6, 7, 9, 11 and 14 were immunodominant and induced interleukin (IL)-10. However, at BOS, the response switched to pep1, 4 and 5 and induced interferon (IFN)-γ and IL-17 responses, but not IL-10. The T helper (Th) phenotype switch is accompanied by decreased frequency of regulatory T cells (T(regs) ) in the lavage. LTx recipients with antibodies to α1(V) also demonstrated increased matrix metalloproteinase (MMP) activation with decreased MMP inhibitor, tissue inhibitor of metalloproteinase (TIMP), suggesting that MMP activation may play a role in the exposure of new Col-V antigenic epitopes. We conclude that a shift in immunodominance of self-antigenic determinants of Col-V results in induction of IFN-γ and IL-17 with loss of tolerance leading to autoimmunity to Col-V, which leads to chronic lung allograft rejection.
Collapse
Affiliation(s)
- V Tiriveedhi
- Department of Surgery, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Real-time monitoring of matrix metalloproteinase-9 collagenolytic activity with a surface plasmon resonance biosensor. Anal Biochem 2011; 419:53-60. [DOI: 10.1016/j.ab.2011.07.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 07/23/2011] [Accepted: 07/25/2011] [Indexed: 11/18/2022]
|
38
|
Robichaud TK, Steffensen B, Fields GB. Exosite interactions impact matrix metalloproteinase collagen specificities. J Biol Chem 2011; 286:37535-42. [PMID: 21896477 DOI: 10.1074/jbc.m111.273391] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Members of the matrix metalloproteinase (MMP) family selectively cleave collagens in vivo. However, the substrate structural determinants that facilitate interaction with specific MMPs are not well defined. We hypothesized that type I-III collagen sequences located N- or C-terminal to the physiological cleavage site mediate substrate selectivity among MMP-1, MMP-2, MMP-8, MMP-13, and MMP-14/membrane-type 1 (MT1)-MMP. The enzyme kinetics for hydrolysis of three fluorogenic triple-helical peptides (fTHPs) was evaluated herein. The first fTHP contained consensus residues 769-783 from type I-III collagens, the second inserted α1(II) collagen residues 763-768 N-terminal to the consensus sequence, and the third inserted α1(II) collagen residues 784-792 C-terminal to the consensus sequence. Our analyses showed that insertion of the C-terminal residues significantly increased k(cat)/K(m) and k(cat) for MMP-1. MMP-13 showed the opposite behavior with a decreased k(cat)/K(m) and k(cat) and a greatly improved K(m) in response to the C-terminal residues. Insertion of the N-terminal residues enhanced k(cat)/K(m) and k(cat) for MMP-8 and MT1-MMP. For MMP-2, the C-terminal residues enhanced K(m) and dramatically decreased k(cat), resulting in a decrease in the overall activity. These changes in activities and kinetic parameters represented the collagen preferences of MMP-8, MMP-13, and MT1-MMP well. Thus, interactions with secondary binding sites (exosites) helped direct the specificity of these enzymes. However, MMP-1 collagen preferences were not recapitulated by the fTHP studies. The preference of MMP-1 for type III collagen appears to be primarily based on the flexibility of the hydrolysis site of type III collagen compared with types I and II. Further characterization of exosite determinants that govern interactions of MMPs with collagenous substrates should aid the development of pharmacotherapeutics that target individual MMPs.
Collapse
Affiliation(s)
- Trista K Robichaud
- Department of Periodontics, University of Texas Health Science Center, San Antonio, Texas 78229, USA
| | | | | |
Collapse
|
39
|
Neujahr DC, Larsen CP. Regulatory T cells in lung transplantation--an emerging concept. Semin Immunopathol 2011; 33:117-27. [PMID: 21424593 PMCID: PMC3395059 DOI: 10.1007/s00281-011-0253-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Accepted: 01/13/2011] [Indexed: 01/11/2023]
Abstract
Lung transplantation represents an option for patients with a variety of end-stage lung diseases. While surgical advances have led to improvements in short-term survival, long-term survival is limited by chronic rejection termed bronchiolitis obliterans syndrome (BOS). A growing body of work is devoted to determining why some patients develop BOS. One avenue of interest that has emerged recently is the role that regulatory T cells (Tregs) may have in protection from BOS. In this review, we will discuss the evidence that Tregs are relevant to outcomes following transplant. We will discuss the relevant animal models, in vitro assays, and human observational studies that support a role for Tregs. We will also explore the interplay between injurious T cells such as Th17 cells and Tregs as well as the effect that additional cell types and chemokines have on the balance between inflammation and regulation. Finally, we will review emerging therapies which may harness the ability of Tregs to lessen the effects of BOS.
Collapse
Affiliation(s)
- David C Neujahr
- Emory University Department of Medicine, Emory University Hospital, Atlanta, GA 30322, USA.
| | | |
Collapse
|
40
|
Giricz O, Lauer JL, Fields GB. Comparison of metalloproteinase protein and activity profiling. Anal Biochem 2011; 409:37-45. [PMID: 20920458 PMCID: PMC3298814 DOI: 10.1016/j.ab.2010.09.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Revised: 09/02/2010] [Accepted: 09/25/2010] [Indexed: 12/23/2022]
Abstract
Proteolytic enzymes play fundamental roles in many biological processes. Members of the matrix metalloproteinase (MMP) family have been shown to take part in processes crucial in disease progression. The current study used the ExcelArray Human MMP/TIMP Array to quantify MMP and tissue inhibitor of metalloproteinase (TIMP) production in the lysates and media of 14 cancer cell lines and 1 normal cell line. The overall patterns were very similar in terms of which MMPs and TIMPs were secreted in the media versus associated with the cells in the individual samples. However, more MMP was found in the media (in both amount and variety). TIMP-1 was produced in all cell lines. MMP activity assays with three different fluorescence resonance energy transfer (FRET) substrates were then used to determine whether protein production correlated with function for the WM-266-4 and BJ cell lines. Metalloproteinase activity was observed for both cell lines with a general MMP substrate (Knight SSP), consistent with protein production data. However, although both cell lines promoted the hydrolysis of a more selective MMP substrate (NFF-3), metalloproteinase activity was confirmed only in the BJ cell line. The use of inhibitors to confirm metalloproteinase activities pointed to the strengths and weaknesses of in situ FRET substrate assays.
Collapse
Affiliation(s)
- Orsi Giricz
- Department of Chemistry & Biochemistry, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431-0991
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
| | - Janelle L. Lauer
- Department of Biochemistry, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229
- Department of Molecular Therapeutics, Scripps Florida, 130 Scripps Way, 2A2, Jupiter, FL 33458
| | - Gregg B. Fields
- Department of Biochemistry, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229
| |
Collapse
|
41
|
Liang X, Arunima A, Zhao Y, Bhaskaran R, Shende A, Byrne TS, Fleeks J, Palmier MO, Van Doren SR. Apparent tradeoff of higher activity in MMP-12 for enhanced stability and flexibility in MMP-3. Biophys J 2010; 99:273-83. [PMID: 20655856 DOI: 10.1016/j.bpj.2010.04.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 03/15/2010] [Accepted: 04/01/2010] [Indexed: 12/17/2022] Open
Abstract
The greater activity of MMP-12 than MMP-3 toward substrates from protein fibrils has been quantified. Why is MMP-12 the more active protease? We looked for behaviors associated with the higher activity of MMP-12 than MMP-3, using nuclear magnetic resonance to monitor backbone dynamics and residue-specific stabilities of their catalytic domain. The proteolytic activities are likely to play important roles in inflammatory diseases of arteries, lungs, joints, and intestines. Nuclear magnetic resonance line broadening indicates that regions surrounding the active sites of both proteases sample conformational substates within milliseconds. The more extensive line broadening in MMP-3 suggests greater sampling of conformational substates, affecting the full length of helix B and beta-strand IV forming the active site, and more remote sites. This could suggest more excursions to functionally incompetent substates. MMP-3 also has enhanced subnanosecond fluctuations in helix A, in the beta-hairpin of strands IV and V, and before and including helix C. Hydrogen exchange protection in the EX2 regime suggests that MMP-3 possesses 2.8 kcal/mol higher folding stability than MMP-12(E219A). The beta-sheet of MMP-3 appears to be stabilized still more. The higher stability of MMP-3 relative to MMP-12 coincides with the former's considerably lower proteolytic activity. This relationship is consistent with the hypothesis that enzymes often trade stability for higher activity.
Collapse
Affiliation(s)
- Xiangyang Liang
- Department of Biochemistry, University of Missouri, Columbia, Missouri, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Xiao J, Addabbo RM, Lauer JL, Fields GB, Baum J. Local conformation and dynamics of isoleucine in the collagenase cleavage site provide a recognition signal for matrix metalloproteinases. J Biol Chem 2010; 285:34181-90. [PMID: 20679339 DOI: 10.1074/jbc.m110.128355] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The mechanism by which enzymes recognize the "uniform" collagen triple helix is not well understood. Matrix metalloproteinases (MMPs) cleave collagen after the Gly residue of the triplet sequence Gly∼[Ile/Leu]-[Ala/Leu] at a single, unique, position along the peptide chain. Sequence analysis of types I-III collagen has revealed a 5-triplet sequence pattern in which the natural cleavage triplets are always flanked by a specific distribution of imino acids. NMR and MMP kinetic studies of a series of homotrimer peptides that model type III collagen have been performed to correlate conformation and dynamics at, and near, the cleavage site to collagenolytic activity. A peptide that models the natural cleavage site is significantly more active than a peptide that models a potential but non-cleavable site just 2-triplets away and NMR studies show clearly that the Ile in the leading chain of the cleavage peptide is more exposed to solvent and less locally stable than the Ile in the middle and lagging chains. We propose that the unique local instability of Ile at the cleavage site in part arises from the placement of the conserved Pro at the P(3) subsite. NMR studies of peptides with Pro substitutions indicate that the local dynamics of the three chains are directly modulated by their proximity to Pro. Correlation of peptide activity to NMR data shows that a single locally unstable chain at the cleavage site, rather than two or three labile chains, is more favorable for cleavage by MMP-1 and may be the determining factor for collagen recognition.
Collapse
Affiliation(s)
- Jianxi Xiao
- Department of Chemistry and Chemical Biology, BIOMAPS Institute, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, USA
| | | | | | | | | |
Collapse
|
43
|
Enhanced proteolytic degradation of molecularly engineered PEG hydrogels in response to MMP-1 and MMP-2. Biomaterials 2010; 31:7836-45. [PMID: 20667588 DOI: 10.1016/j.biomaterials.2010.06.061] [Citation(s) in RCA: 419] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Accepted: 06/30/2010] [Indexed: 12/12/2022]
Abstract
Bioactive hydrogels formed by Michael-type addition reactions of end-functionalized poly(ethylene glycol) macromers with cysteine-containing peptides have been described as extracellular matrix mimetics and tissue engineering scaffolds. Although these materials have shown favorable behavior in vivo in tissue repair, we sought to develop materials formulations that would be more rapidly responsive to cell-induced enzymatic remodeling. In this study, protease-sensitive peptides that have increased k(cat) values were characterized and evaluated for their effects on gel degradability. Biochemical properties for soluble peptides and hydrogels were examined for matrix metalloproteinase (MMP)-1 and MMP-2. The most efficient peptide substrates in some cases overlap and in other cases differ between the two enzymes tested, and a range of k(cat) values was obtained. For each enzyme, hydrogels formed using the peptides with higher k(cat) values degraded faster than a reference with lower k(cat). Fibroblasts showed increased cell spreading and proliferation when cultured in 3D hydrogels with faster degrading peptides, and more cell invasion from aortic ring segments embedded in the hydrogels was observed. These faster degrading gels should provide matrices that are easier for cells to remodel and lead to increased cellular infiltration and potentially more robust healing in vivo.
Collapse
|
44
|
Palmier MO, Fulcher YG, Bhaskaran R, Duong VQ, Fields GB, Van Doren SR. NMR and bioinformatics discovery of exosites that tune metalloelastase specificity for solubilized elastin and collagen triple helices. J Biol Chem 2010; 285:30918-30. [PMID: 20663866 DOI: 10.1074/jbc.m110.136903] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The catalytic domain of metalloelastase (matrix metalloproteinase-12 or MMP-12) is unique among MMPs in exerting high proteolytic activity upon fibrils that resist hydrolysis, especially elastin from lungs afflicted with chronic obstructive pulmonary disease or arteries with aneurysms. How does the MMP-12 catalytic domain achieve this specificity? NMR interface mapping suggests that α-elastin species cover the primed subsites, a strip across the β-sheet from β-strand IV to the II-III loop, and a broad bowl from helix A to helix C. The many contacts may account for the comparatively high affinity, as well as embedding of MMP-12 in damaged elastin fibrils in vivo. We developed a strategy called BINDSIght, for bioinformatics and NMR discovery of specificity of interactions, to evaluate MMP-12 specificity without a structure of a complex. BINDSIght integration of the interface mapping with other ambiguous information from sequences guided choice mutations in binding regions nearer the active site. Single substitutions at each of ten locations impair specific activity toward solubilized elastin. Five of them impair release of peptides from intact elastin fibrils. Eight lesions also impair specific activity toward triple helices from collagen IV or V. Eight sites map to the "primed" side in the III-IV, V-B, and S1' specificity loops. Two map to the "unprimed" side in the IV-V and B-C loops. The ten key residues circumscribe the catalytic cleft, form an exosite, and are distinctive features available for targeting by new diagnostics or therapeutics.
Collapse
Affiliation(s)
- Mark O Palmier
- Department of Biochemistry, University of Missouri, Columbia, Missouri 65211, USA
| | | | | | | | | | | |
Collapse
|
45
|
Singh NK, Quyen DV, Kundumani-Sridharan V, Brooks PC, Rao GN. AP-1 (Fra-1/c-Jun)-mediated induction of expression of matrix metalloproteinase-2 is required for 15S-hydroxyeicosatetraenoic acid-induced angiogenesis. J Biol Chem 2010; 285:16830-43. [PMID: 20353950 DOI: 10.1074/jbc.m110.106187] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To understand the involvement of matrix metalloproteinases (MMPs) in 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE)-induced angiogenesis, we have studied the role of MMP-2. 15(S)-HETE induced MMP-2 expression and activity in a time-dependent manner in human dermal microvascular endothelial cells (HDMVECs). Inhibition of MMP-2 activity or depletion of its levels attenuated 15(S)-HETE-induced HDMVEC migration, tube formation, and Matrigel plug angiogenesis. 15(S)-HETE also induced Fra-1 and c-Jun expression in a Rac1-MEK1-JNK1-dependent manner. In addition, 15(S)-HETE-induced MMP-2 expression and activity were mediated by Rac1-MEK1-JNK1-dependent activation of AP-1 (Fra-1/c-Jun). Cloning and site-directed mutagenesis of MMP-2 promoter revealed that AP-1 site proximal to the transcriptional start site is required for 15(S)-HETE-induced MMP-2 expression, and Fra-1 and c-Jun are the essential components of AP-1 that bind to MMP-2 promoter in response to 15(S)-HETE. Hind limb ischemia led to an increase in MEK1 and JNK1 activation and Fra-1, c-Jun, and MMP-2 expression resulting in enhanced neovascularization and recovery of blood perfusion in wild-type mice as compared with 12/15-Lox(-/-) mice. Together, these results provide the first direct evidence for a role of 12/15-Lox-12/15(S)-HETE axis in the regulation of ischemia-induced angiogenesis.
Collapse
Affiliation(s)
- Nikhlesh K Singh
- Department of Physiology, the University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | | | | | | | |
Collapse
|
46
|
Abstract
Triple-helical peptides (THPs) have been utilized as collagen models since the 1960s. The original focus for THP-based research was to unravel the structural determinants of collagen. In the last two decades, virtually all aspects of collagen structural biochemistry have been explored with THP models. More specifically, secondary amino acid analogs have been incorporated into THPs to more fully understand the forces that stabilize triple-helical structure. Heterotrimeric THPs have been utilized to better appreciate the contributions of chain sequence diversity on collagen function. The role of collagen as a cell signaling protein has been dissected using THPs that represent ligands for specific receptors. The mechanisms of collagenolysis have been investigated using THP substrates and inhibitors. Finally, THPs have been developed for biomaterial applications. These aspects of THP-based research are overviewed herein.
Collapse
Affiliation(s)
- Gregg B Fields
- University of Texas Health Science Center, Department of Biochemistry, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
47
|
Abstract
The recognition that the successful clinical use of MMP inhibitors will require quantitative correlation of MMP activity with disease type, and to disease progression, has stimulated intensive effort toward the development of sensitive assay methods, improved analytical methods for the determination of the structural profile for MMP-sub-type inhibition, and the development of new methods for the determination - in both quantitative and qualitative terms - of MMP activity. This chapter reviews recent progress toward these objectives, with particular emphasis on the quantitative and qualitative profiling of MMP activity in cells and tissues. Quantitative determination of MMP activity is made from the concentration of the MMP from the tissue, using immobilization of a broad-spectrum MMP inhibitor on a chromatography resin. Active MMP, to the exclusion of MMP zymogens and endogenous TIMP-inhibited MMPs, is retained on the column. Characterization of the MMP sub-type(s) follows from appropriate analysis of the active MMP eluted from the resin. Qualitative determination of MMP involvement in disease can be made using an MMP sub-type-selective inhibitor. The proof of principle, with respect to this qualitative determination of the disease involvement of the gelatinase MMP-2 and MMP-9 sub-types, is provided by the class of thiirane-based MMP mechanism-based inhibitors (SB-3CT as the prototype). Positive outcomes in animal models of disease having MMP-2 and/or -9 dependency follow administration of this MMP inhibitor, whereas this inhibitor is inactive in disease models where other MMPs (such as MMP-14) are involved.
Collapse
Affiliation(s)
- Jed F Fisher
- Department of Chemistry and Biochemistry, Walther Cancer Research Center, University of Notre Dame, Notre Dame, IN, USA
| | | |
Collapse
|
48
|
Abstract
A continuous assay method, such as the one that utilizes an increase in fluorescence upon hydrolysis, allows for rapid and convenient kinetic evaluation of proteases. To better understand MMP behaviors and to aid in the design of MMP inhibitors, a variety of sequence specificity, phage display, and combinatorial chemistry studies have been performed. Results of these studies have been valuable for defining the differences in MMPs and for creating quenched fluorescent substrates that utilize fluorescence resonance energy transfer (FRET)/intramolecular fluorescence energy transfer (IFET). FRET triple-helical substrates have been constructed to examine the collagenolytic activity of MMP family members. The present chapter provides an overview of MMP and related FRET substrates and describes how to construct and utilize these substrates.
Collapse
|
49
|
Banerjee J, Hanson AJ, Gadam B, Elegbede AI, Tobwala S, Ganguly B, Wagh AV, Muhonen WW, Law B, Shabb JB, Srivastava DK, Mallik S. Release of liposomal contents by cell-secreted matrix metalloproteinase-9. Bioconjug Chem 2009; 20:1332-9. [PMID: 19601658 DOI: 10.1021/bc9000646] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Liposomes have been widely used as a drug delivery vehicle, and currently, more than 10 liposomal formulations are approved by the Food and Drug Administration for clinical use. However, upon targeting, the release of the liposome-encapsulated contents is usually slow. We have recently demonstrated that contents from appropriately formulated liposomes can be rapidly released by the cancer-associated enzyme matrix metalloproteinase-9 (MMP-9). Herein, we report our detailed studies to optimize the liposomal formulations. By properly selecting the lipopeptide, the major lipid component, and their relative amounts, we demonstrate that the contents are rapidly released in the presence of cancer-associated levels of recombinant human MMP-9. We observed that the degree of lipid mismatch between the lipopepides and the major lipid component profoundly affects the release profiles from the liposomes. By utilizing the optimized liposomal formulations, we also demonstrate that cancer cells (HT-29) which secrete low levels of MMP-9 failed to release a significant amount of the liposomal contents. Metastatic cancer cells (MCF7) secreting high levels of the enzyme rapidly release the encapsulated contents from the liposomes.
Collapse
Affiliation(s)
- Jayati Banerjee
- Department of Pharmaceutical Sciences, Department of Chemistry, Biochemistry and Molecular Biology, North Dakota State University, Fargo, North Dakota 58108, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Jastrzebska B, Lebel R, Therriault H, McIntyre JO, Escher E, Guérin B, Paquette B, Neugebauer WA, Lepage M. New enzyme-activated solubility-switchable contrast agent for magnetic resonance imaging: from synthesis to in vivo imaging. J Med Chem 2009; 52:1576-81. [PMID: 19228016 DOI: 10.1021/jm801411h] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We designed and synthesized a novel contrast agent (CA) to image the activity of matrix metalloproteinase-2 (MMP-2) in a tumor, noninvasively using magnetic resonance imaging (MRI). We exploited the concept of solubility-switchable CAs in the design of a protease-modulated CA (PCA), referred to as PCA2-switch. This PCA contains a paramagnetic gadolinium chelate (Gd-DOTA), which was attached to the N-terminus of a MMP-2 cleavable peptide sequence via a hydrophobic chain. The aqueous solubility of the CA depends on the presence of a polyethylene glycol chain (PEG) on the C-terminus of the peptide. Upon proteolytic cleavage of the peptide by MMP-2, the PEG chain is detached from the CA, which becomes less water soluble. This compound and control compounds were successfully tested in an animal model bearing two tumors with different levels of MMP-2 activity.
Collapse
Affiliation(s)
- Beata Jastrzebska
- Centre d'Imagerie Moleculaire de Sherbrooke and Department of Nuclear Medicine and Radiobiology, Universite de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|