1
|
Dari S, O'dea RD, Fadai NT. Understanding the regulation of chronic wounds by tissue inhibitors of matrix metalloproteinases through mathematical modelling. J Theor Biol 2025; 604:112083. [PMID: 40020775 DOI: 10.1016/j.jtbi.2025.112083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 02/21/2025] [Accepted: 02/21/2025] [Indexed: 03/03/2025]
Abstract
Understanding the biochemistry and pharmacodynamics of chronic wounds is of key importance, due to the millions of people in the UK affected and the significant cost to the NHS. Chronic wounds are characterised by elevated concentrations of matrix metalloproteinases (MMPs) that destroy the surrounding extracellular matrix (ECM). However, fibroblasts can produce tissue inhibitors of MMPs (TIMPs) in order to regulate wound healing. Therefore, the role of TIMPs in both acute and chronic wounds needs to be properly understood in order to develop therapeutic treatments. In this work, we propose a reaction-diffusion system of four partial differential equations that describe the interaction of the ECM, fibroblasts, MMPs, and TIMPs in a wound. We observe that, subject to parameter sets corresponding to both acute and chronic wound healing, this mathematical model gives rise to travelling wave solutions. Using bifurcation analysis, we demonstrate that excessive degradation of the ECM results in the emergence of chronic wounds, and the reversal of these chronic wounds is prohibited for lower TIMP production values. These results are replicated within a simplified model obtained via a parameter sensitivity analysis. This model is further extended to more realistic spatial domains where we demonstrate the effectiveness of a therapeutic hydrogel containing TIMPs as a treatment for chronic wounds.
Collapse
Affiliation(s)
- Sonia Dari
- School of Mathematical Sciences, University of Nottingham, Nottingham, NG7 2RD, UK.
| | - Reuben D O'dea
- School of Mathematical Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Nabil T Fadai
- School of Mathematical Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| |
Collapse
|
2
|
Dalvi S, Roll M, Chatterjee A, Kumar LK, Bhogavalli A, Foley N, Arduino C, Spencer W, Reuben-Thomas C, Ortolan D, Pébay A, Bharti K, Anand-Apte B, Singh R. Human iPSC-based disease modeling studies identify a common mechanistic defect and potential therapies for AMD and related macular dystrophies. Dev Cell 2024; 59:3290-3305.e9. [PMID: 39362220 PMCID: PMC11652237 DOI: 10.1016/j.devcel.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 04/17/2024] [Accepted: 09/05/2024] [Indexed: 10/05/2024]
Abstract
Age-related macular degeneration (AMD) and related macular dystrophies (MDs) primarily affect the retinal pigment epithelium (RPE) in the eye. A hallmark of AMD/MDs that drives later-stage pathologies is drusen. Drusen are sub-RPE lipid-protein-rich extracellular deposits, but how drusen forms and accumulates is not known. We utilized human induced pluripotent stem cell (iPSC)-derived RPE from patients with AMD and three distinct MDs to demonstrate that reduced activity of RPE-secreted matrix metalloproteinase 2 (MMP2) contributes to drusen in multiple maculopathies in a genotype-agnostic manner by instigating sterile inflammation and impaired lipid homeostasis via damage-associated molecular pattern molecule (DAMP)-mediated activation of receptor for advanced glycation end-products (RAGE) and increased secretory phospholipase 2-IIA (sPLA2-IIA) levels. Therapeutically, RPE-specific MMP2 supplementation, RAGE-antagonistic peptide, and a small molecule inhibitor of sPLA2-IIA ameliorated drusen accumulation in AMD/MD iPSC-RPE. Ultimately, this study defines a causal role of the MMP2-DAMP-RAGE-sPLA2-IIA axis in AMD/MDs.
Collapse
Affiliation(s)
- Sonal Dalvi
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Michael Roll
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Amit Chatterjee
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Lal Krishan Kumar
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Akshita Bhogavalli
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Nathaniel Foley
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Cesar Arduino
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Whitney Spencer
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Cheyenne Reuben-Thomas
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA
| | - Davide Ortolan
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Alice Pébay
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Bela Anand-Apte
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Ruchira Singh
- Department of Ophthalmology, University of Rochester, Rochester, NY 14620, USA; Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; Center for Visual Science, University of Rochester, Rochester, NY 14620, USA; UR Stem Cell and Regenerative Medicine Center, Rochester, NY 14620, USA.
| |
Collapse
|
3
|
Aldaghi N, Kamalabadi-Farahani M, Alizadeh M, Salehi M. Doxycycline-loaded carboxymethyl cellulose/sodium alginate/gelatin hydrogel: An approach for enhancing pressure ulcer healing in a rat model. J Biomed Mater Res A 2024; 112:2289-2300. [PMID: 39019482 DOI: 10.1002/jbm.a.37778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/15/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024]
Abstract
Pressure ulcers, or bedsores, are created by areas of the skin under prolonged pressure and can lead to skin and underlying tissue damage. The present study evaluated the effects of carboxymethyl cellulose/sodium alginate/gelatin (CMC/Alg/Gel) hydrogel containing doxycycline (DOX) on improving the healing process of pressure ulcers. The magnet was used to apply pressure on the dorsum skin rat to induce a pressure ulcer model. Then sterile gauze, CMC/Alg/Gel, and CMC/Alg/Gel/1% w/v DOX hydrogels were used to cover the wounds. Blood compatibility, weight loss, cytocompatibility, drug release rate, cell viability, wound closure, and re-epithelialization were evaluated in all animals on the 14th day after treatment. In vivo results and histopathological evaluation showed 56.66% wound closure and the highest re-epithelialization in the CMC/Alg/Gel/1% w/v DOX hydrogel group (14 days after treatment). Furthermore, real-time PCR results indicated that the hydrogel containing DOX significantly decreased the expression of the MMP family consisting of MMP2 and MMP9 mRNA and also increased the expression of vascular endothelial growth factor VEGF mRNA. This study suggested that the addition of DOX, an antibiotic and MMP inhibitor, to hydrogels may be effective in the healing process of pressure ulcers.
Collapse
Affiliation(s)
- Niloofar Aldaghi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohammad Kamalabadi-Farahani
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Morteza Alizadeh
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Majid Salehi
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Health Technology Incubator Center, Shahroud University of Medical Sciences, Shahroud, Iran
- Sexual Health and Fertility Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
4
|
Chang HF, Cheng JY. Glioblastoma U-87 cell electrotaxis is hindered by doxycycline with a concomitant reduction in the matrix metallopeptidase-9 expression. Biochem Biophys Rep 2024; 38:101690. [PMID: 38571555 PMCID: PMC10987802 DOI: 10.1016/j.bbrep.2024.101690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
Electric fields (EF) play an essential role in cancer cell migration. Numerous cancer cell types exhibit electrotaxis under direct current electric fields (dcEF) of physiological electric field strength (EFs). This study investigated the effects of doxycycline on the electrotactic responses of U87 cells. After EF stimulation, U87 cells migrated toward the cathode, whereas doxycycline-treated U87 cells exhibited enhanced cell mobility but hindered cathodal migration. We further investigated the expression of the metastasis-correlated proteins matrix metallopeptidase-2 (MMP-2) and MMP-9 in U87 cells. The levels of MMP-2 in the cells were not altered under EF or doxycycline stimulation. In contrast, the EF stimulation greatly enhanced the levels of MMP-9 and then repressed in doxycycline-cotreated cells, accompanied by reduced cathodal migration. Our results demonstrated that an antibiotic at a non-toxic concentration could suppress the enhanced cell migration accelerated by EF of physiological strength. This finding may be applied as an anti-metastatic treatment for cancers.
Collapse
Affiliation(s)
- Hui-Fang Chang
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Ji-Yen Cheng
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Mechanical and Mechatronic Engineering, National Taiwan Ocean University, Keelung, Taiwan
- College of Engineering, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
5
|
Barakat N, Jangir H, Gallo L, Grillo M, Guo X, Hickman J. Inhibition of Metalloproteinases Extends Longevity and Function of In Vitro Human iPSC-Derived Skeletal Muscle. Biomedicines 2024; 12:856. [PMID: 38672210 PMCID: PMC11047953 DOI: 10.3390/biomedicines12040856] [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: 02/26/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
In vitro culture longevity has long been a concern for disease modeling and drug testing when using contractable cells. The dynamic nature of certain cells, such as skeletal muscle, contributes to cell surface release, which limits the system's ability to conduct long-term studies. This study hypothesized that regulating the extracellular matrix (ECM) dynamics should be able to prolong cell attachment on a culture surface. Human induced pluripotent stem cell (iPSC)-derived skeletal muscle (SKM) culture was utilized to test this hypothesis due to its forceful contractions in mature muscle culture, which can cause cell detachment. By specifically inhibiting matrix metalloproteinases (MMPs) that work to digest components of the ECM, it was shown that the SKM culture remained adhered for longer periods of time, up to 80 days. Functional testing of myofibers indicated that cells treated with the MMP inhibitors, tempol, and doxycycline, displayed a significantly reduced fatigue index, although the fidelity was not affected, while those treated with the MMP inducer, PMA, indicated a premature detachment and increased fatigue index. The MMP-modulating activity by the inhibitors and inducer was further validated by gel zymography analysis, where the MMP inhibitor showed minimally active MMPs, while the inducer-treated cells indicated high MMP activity. These data support the hypotheses that regulating the ECM dynamics can help maximize in vitro myotube longevity. This proof-of-principle strategy would benefit the modeling of diseases that require a long time to develop and the evaluation of chronic effects of potential therapeutics.
Collapse
Affiliation(s)
- Natali Barakat
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (N.B.); (H.J.); (L.G.); (M.G.); (X.G.)
- Department of Chemistry, University of Central Florida, Orlando, FL 32828, USA
| | - Himanshi Jangir
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (N.B.); (H.J.); (L.G.); (M.G.); (X.G.)
| | - Leandro Gallo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (N.B.); (H.J.); (L.G.); (M.G.); (X.G.)
| | - Marcella Grillo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (N.B.); (H.J.); (L.G.); (M.G.); (X.G.)
| | - Xiufang Guo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (N.B.); (H.J.); (L.G.); (M.G.); (X.G.)
| | - James Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (N.B.); (H.J.); (L.G.); (M.G.); (X.G.)
- Department of Chemistry, University of Central Florida, Orlando, FL 32828, USA
| |
Collapse
|
6
|
Du P, Hou Y, Su C, Gao J, Yang Y, Zhang J, Cui X, Tang J. The future for the therapeutics of abdominal aortic aneurysm: engineered nanoparticles drug delivery for abdominal aortic aneurysm. Front Bioeng Biotechnol 2024; 11:1324406. [PMID: 38249799 PMCID: PMC10796665 DOI: 10.3389/fbioe.2023.1324406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a severe cardiovascular disease with a high mortality rate. Several screening and diagnostic methods have been developed for AAA early diagnosis. Open surgery and endovascular aortic repair (EVAR) are clinically available for patients who meet the indications for surgery. However, for non-surgical patients, limited drugs exist to inhibit or reverse the progression of aneurysms due to the complex pathogenesis and biological structure of AAA, failing to accumulate precisely on the lesion to achieve sufficient concentrations. The recently developed nanotechnology offers a new strategy to address this problem by developing drug-carrying nanoparticles with enhanced water solubility and targeting capacity, prolonged duration, and reduced side effects. Despite the rising popularity, limited literature is available to highlight the progression of the field. Herein, in this review, we first discuss the pathogenesis of AAA, the methods of diagnosis and treatment that have been applied clinically, followed by the review of research progressions of constructing different drug-loaded nanoparticles for AAA treatment using engineered nanoparticles. In addition, the feasibility of extracellular vesicles (EVs) and EVs-based nanotechnology for AAA treatment in recent years are highlighted, together with the future perspective. We hope this review will provide a clear picture for the scientists and clinicians to find a new solution for AAA clinical management.
Collapse
Affiliation(s)
- Pengchong Du
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Yachen Hou
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Chang Su
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Jiamin Gao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Yu Yang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Jinying Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Xiaolin Cui
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Junnan Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| |
Collapse
|
7
|
Hartley B, Bassiouni W, Schulz R, Julien O. The roles of intracellular proteolysis in cardiac ischemia-reperfusion injury. Basic Res Cardiol 2023; 118:38. [PMID: 37768438 DOI: 10.1007/s00395-023-01007-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023]
Abstract
Ischemic heart disease remains a leading cause of human mortality worldwide. One form of ischemic heart disease is ischemia-reperfusion injury caused by the reintroduction of blood supply to ischemic cardiac muscle. The short and long-term damage that occurs due to ischemia-reperfusion injury is partly due to the proteolysis of diverse protein substrates inside and outside of cardiomyocytes. Ischemia-reperfusion activates several diverse intracellular proteases, including, but not limited to, matrix metalloproteinases, calpains, cathepsins, and caspases. This review will focus on the biological roles, intracellular localization, proteolytic targets, and inhibitors of these proteases in cardiomyocytes following ischemia-reperfusion injury. Recognition of the intracellular function of each of these proteases includes defining their activation, proteolytic targets, and their inhibitors during myocardial ischemia-reperfusion injury. This review is a step toward a better understanding of protease activation and involvement in ischemic heart disease and developing new therapeutic strategies for its treatment.
Collapse
Affiliation(s)
- Bridgette Hartley
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Wesam Bassiouni
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - Richard Schulz
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada.
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada.
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada.
| | - Olivier Julien
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
8
|
Atkinson G, Bianco R, Di Gregoli K, Johnson JL. The contribution of matrix metalloproteinases and their inhibitors to the development, progression, and rupture of abdominal aortic aneurysms. Front Cardiovasc Med 2023; 10:1248561. [PMID: 37799778 PMCID: PMC10549934 DOI: 10.3389/fcvm.2023.1248561] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/07/2023] [Indexed: 10/07/2023] Open
Abstract
Abdominal aortic aneurysms (AAAs) account for up to 8% of deaths in men aged 65 years and over and 2.2% of women. Patients with AAAs often have atherosclerosis, and intimal atherosclerosis is generally present in AAAs. Accordingly, AAAs are considered a form of atherosclerosis and are frequently referred to as atherosclerotic aneurysms. Pathological observations advocate inflammatory cell infiltration alongside adverse extracellular matrix degradation as key contributing factors to the formation of human atherosclerotic AAAs. Therefore, macrophage production of proteolytic enzymes is deemed responsible for the damaging loss of ECM proteins, especially elastin and fibrillar collagens, which characterise AAA progression and rupture. Matrix metalloproteinases (MMPs) and their regulation by tissue inhibitors metalloproteinases (TIMPs) can orchestrate not only ECM remodelling, but also moderate the proliferation, migration, and apoptosis of resident aortic cells, alongside the recruitment and subsequent behaviour of inflammatory cells. Accordingly, MMPs are thought to play a central regulatory role in the development, progression, and eventual rupture of abdominal aortic aneurysms (AAAs). Together, clinical and animal studies have shed light on the complex and often diverse effects MMPs and TIMPs impart during the development of AAAs. This dichotomy is underlined from evidence utilising broad-spectrum MMP inhibition in animal models and clinical trials which have failed to provide consistent protection from AAA progression, although more encouraging results have been observed through deployment of selective inhibitors. This review provides a summary of the supporting evidence connecting the contribution of individual MMPs to AAA development, progression, and eventual rupture. Topics discussed include structural, functional, and cell-specific diversity of MMP members; evidence from animal models of AAA and comparisons with findings in humans; the dual role of MMPs and the requirement to selectively target individual MMPs; and the advances in identifying aberrant MMP activity. As evidenced, our developing understanding of the multifaceted roles individual MMPs perform during the progression and rupture of AAAs, should motivate clinical trials assessing the therapeutic potential of selective MMP inhibitors, which could restrict AAA-related morbidity and mortality worldwide.
Collapse
Affiliation(s)
| | | | | | - Jason L. Johnson
- Laboratory of Cardiovascular Pathology, Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
9
|
Dinte E, Muntean DM, Andrei V, Boșca BA, Dudescu CM, Barbu-Tudoran L, Borodi G, Andrei S, Gal AF, Rus V, Gherman LM, Cadar O, Barabas R, Niculae M, Ilea A. In Vitro and In Vivo Characterisation of a Mucoadhesive Buccal Film Loaded with Doxycycline Hyclate for Topical Application in Periodontitis. Pharmaceutics 2023; 15:pharmaceutics15020580. [PMID: 36839899 PMCID: PMC9963859 DOI: 10.3390/pharmaceutics15020580] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Mucoadhesive films loaded with doxycycline hyclate (Doxy Hyc), consisting of mixtures of hydroxypropylmethyl cellulose (HPMC) E3, K4 and polyacrylic acid (Carbopol 940), were prepared by casting method, aiming to design a formulation intended for application in the oral cavity. The obtained film formulations exhibited a Doxy Hyc content between 7.52 ± 0.42 and 7.83 ± 0.41%, which had adequate mechanical properties for application in the oral cavity and pH values in the tolerance range. The x-ray diffraction studies highlighted the amorphisation of Doxy Hyc in the preparation process and the antibiotic particles present on the surface of the films, identified in the TEM images, which ensured a burst release effect in the first 15 min of the in vitro dissolution studies, after which Doxy Hyc was released by diffusion, the data presenting a good correlation with the Peppas model, n < 0.5. The formulation F1, consisting of HPMC K4 combined with C940 in a ratio of 5:3, the most performing in vitro, was tested in vivo in experimentally-induced periodontitis and demonstrated its effectiveness in improving the clinical parameters and reducing the salivary levels of matrix metalloproteinase-8 (MMP-8). The prepared Doxy Hyc loaded mucoadhesive buccal film could be used as an adjuvant for the local treatment of periodontitis, ensuring prolonged release of the antibiotic after topical application.
Collapse
Affiliation(s)
- Elena Dinte
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Dana Maria Muntean
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Correspondence:
| | - Vlad Andrei
- Department of Oral Rehabilitation, Faculty of Dentistry, ”Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Bianca Adina Boșca
- Department of Morphological Sciences, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Cristian Mircea Dudescu
- Department of Mechanical Engineering, Faculty of Automotive, Mechatronics and Mechanical Engineering, Technical University of Cluj-Napoca, 400641 Cluj-Napoca, Romania
| | - Lucian Barbu-Tudoran
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Gheorghe Borodi
- National Institute for Research and Development of Isotopic and Molecular Technologies, 400293 Cluj-Napoca, Romania
| | - Sanda Andrei
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | - Adrian Florin Gal
- Department of Cell Biology, Histology and Embryology, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | - Vasile Rus
- Department of Cell Biology, Histology and Embryology, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | - Luciana-Mădălina Gherman
- Experimental Centre of University of Medicine and Pharmacy “Iuliu Hațieganu”, 400349 Cluj-Napoca, Romania
| | - Oana Cadar
- INCDO-INOE 2000, Research Institute for Analytical Instrumentation, 400293 Cluj-Napoca, Romania
| | - Reka Barabas
- Department of Chemistry and Chemical Engineering of Hungarian Line of Study, Faculty of Chemistry and Chemical Engineering, 400028 Cluj-Napoca, Romania
| | - Mihaela Niculae
- Department of Infectious Diseases, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | - Aranka Ilea
- Department of Oral Rehabilitation, Faculty of Dentistry, ”Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
10
|
Escalona RM, Chu S, Kadife E, Kelly JK, Kannourakis G, Findlay JK, Ahmed N. Knock down of TIMP-2 by siRNA and CRISPR/Cas9 mediates diverse cellular reprogramming of metastasis and chemosensitivity in ovarian cancer. Cancer Cell Int 2022; 22:422. [PMID: 36585738 PMCID: PMC9805260 DOI: 10.1186/s12935-022-02838-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The endogenous tissue inhibitor of metalloproteinase-2 (TIMP-2), through its homeostatic action on certain metalloproteinases, plays a vital role in remodelling extracellular matrix (ECM) to facilitate cancer progression. This study investigated the role of TIMP-2 in an ovarian cancer cell line in which the expression of TIMP-2 was reduced by either siRNA or CRISPR/Cas9. METHODS OVCAR5 cells were transiently and stably transfected with either single or pooled TIMP-2 siRNAs (T2-KD cells) or by CRISPR/Cas9 under the influence of two distinct guide RNAs (gRNA1 and gRNA2 cell lines). The expression of different genes was analysed at the mRNA level by quantitative real time PCR (qRT-PCR) and at the protein level by immunofluorescence (IF) and western blot. Proliferation of cells was investigated by 5-Ethynyl-2'-deoxyuridine (EdU) assay or staining with Ki67. Cell migration/invasion was determined by xCELLigence. Cell growth in vitro was determined by 3D spheroid cultures and in vivo by a mouse xenograft model. RESULTS Approximately 70-90% knock down of TIMP-2 expression were confirmed in T2-KD, gRNA1 and gRNA2 OVCAR5 ovarian cancer cells at the protein level. T2-KD, gRNA1 and gRNA2 cells exhibited a significant downregulation of MMP-2 expression, but concurrently a significant upregulation in the expression of membrane bound MMP-14 compared to control and parental cells. Enhanced proliferation and invasion were exhibited in all TIMP-2 knocked down cells but differences in sensitivity to paclitaxel (PTX) treatment were observed, with T2-KD cells and gRNA2 cell line being sensitive, while the gRNA1 cell line was resistant to PTX treatment. In addition, significant differences in the growth of gRNA1 and gRNA2 cell lines were observed in in vitro 3D cultures as well as in an in vivo mouse xenograft model. CONCLUSIONS Our results suggest that the inhibition of TIMP-2 by siRNA and CRISPR/Cas-9 modulate the expression of MMP-2 and MMP-14 and reprogram ovarian cancer cells to facilitate proliferation and invasion. Distinct disparities in in vitro chemosensitivity and growth in 3D culture, and differences in tumour burden and invasion to proximal organs in a mouse model imply that selective suppression of TIMP-2 expression by siRNA or CRISPR/Cas-9 alters important aspects of metastasis and chemosensitivity in ovarian cancer.
Collapse
Affiliation(s)
- Ruth M. Escalona
- grid.1008.90000 0001 2179 088XDepartment of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC 3052 Australia ,grid.1002.30000 0004 1936 7857Centre for Reproductive Health, Hudson Institute of Medical Research and Department of Translational Medicine, Monash University, Clayton, VIC 3168 Australia ,Fiona Elsey Cancer Research Institute, Suites 23, 106-110 Lydiard Street South, Ballarat Technology Park Central, Ballarat, VIC 3350 Australia
| | - Simon Chu
- grid.1002.30000 0004 1936 7857Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research and Department of Translational Medicine, Monash University, Clayton, VIC 3168 Australia
| | - Elif Kadife
- Fiona Elsey Cancer Research Institute, Suites 23, 106-110 Lydiard Street South, Ballarat Technology Park Central, Ballarat, VIC 3350 Australia
| | - Jason K. Kelly
- Fiona Elsey Cancer Research Institute, Suites 23, 106-110 Lydiard Street South, Ballarat Technology Park Central, Ballarat, VIC 3350 Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Suites 23, 106-110 Lydiard Street South, Ballarat Technology Park Central, Ballarat, VIC 3350 Australia ,grid.1040.50000 0001 1091 4859School of Science, Psychology and Sport, Federation University, Mt Helen, VIC 3350 Australia
| | - Jock K. Findlay
- grid.1008.90000 0001 2179 088XDepartment of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC 3052 Australia ,grid.1002.30000 0004 1936 7857Centre for Reproductive Health, Hudson Institute of Medical Research and Department of Translational Medicine, Monash University, Clayton, VIC 3168 Australia
| | - Nuzhat Ahmed
- grid.1008.90000 0001 2179 088XDepartment of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC 3052 Australia ,grid.1002.30000 0004 1936 7857Centre for Reproductive Health, Hudson Institute of Medical Research and Department of Translational Medicine, Monash University, Clayton, VIC 3168 Australia ,Fiona Elsey Cancer Research Institute, Suites 23, 106-110 Lydiard Street South, Ballarat Technology Park Central, Ballarat, VIC 3350 Australia ,grid.1040.50000 0001 1091 4859School of Science, Psychology and Sport, Federation University, Mt Helen, VIC 3350 Australia
| |
Collapse
|
11
|
Li X, Khan D, Rana M, Hänggi D, Muhammad S. Doxycycline Attenuated Ethanol-Induced Inflammaging in Endothelial Cells: Implications in Alcohol-Mediated Vascular Diseases. Antioxidants (Basel) 2022; 11:antiox11122413. [PMID: 36552622 PMCID: PMC9774758 DOI: 10.3390/antiox11122413] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/05/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Excess alcohol consumption is a potential risk factor for cardiovascular diseases and is linked to accelerated aging. Drug discovery to reduce toxic cellular events of alcohol is required. Here, we investigated the effects of ethanol on human umbilical vein endothelial cells (HUVECs) and explored if doxycycline attenuates ethanol-mediated molecular events in endothelial cells. Initially, a drug screening using a panel of 170 drugs was performed, and doxycycline was selected for further experiments. HUVECs were treated with different concentrations (300 mM and 400 mM) of ethanol with or without doxycycline (10 µg/mL). Telomere length was quantified as telomere to single-copy gene (T/S) ratio. Telomere length and the mRNA expression were quantified by qRT-PCR, and protein level was analyzed by Western blot (WB). Ethanol treatment accelerated cellular aging, and doxycycline treatment recovered telomere length. Pathway analysis showed that doxycycline inhibited mTOR and NFκ-B activation. Doxycycline restored the expression of aging-associated proteins, including lamin b1 and DNA repair proteins KU70 and KU80. Doxycycline reduced senescence and senescence-associated secretory phenotype (SASP) in ethanol-treated HUVECs. In conclusion, we report that ethanol-induced inflammation and aging in HUVECs were ameliorated by doxycycline.
Collapse
Affiliation(s)
- Xuanchen Li
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Dilaware Khan
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
- Correspondence: ; Tel.: +49-21181-08782
| | - Majeed Rana
- Department of Oral and Maxillofacial Surgery, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Daniel Hänggi
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Sajjad Muhammad
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
- Department of Neurosurgery, University Hospital Helsinki, Topeliuksenkatu 5, 00260 Helsinki, Finland
| |
Collapse
|
12
|
Weaver LM, Loftin CD, Zhan CG. Development of pharmacotherapies for abdominal aortic aneurysms. Biomed Pharmacother 2022; 153:113340. [PMID: 35780618 PMCID: PMC9514980 DOI: 10.1016/j.biopha.2022.113340] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/13/2022] [Accepted: 06/24/2022] [Indexed: 11/23/2022] Open
Abstract
The cardiovascular field is still searching for a treatment for abdominal aortic aneurysms (AAA). This inflammatory disease often goes undiagnosed until a late stage and associated rupture has a high mortality rate. No pharmacological treatment options are available. Three hallmark factors of AAA pathology include inflammation, extracellular matrix remodeling, and vascular smooth muscle dysfunction. Here we discuss drugs for AAA treatment that have been studied in clinical trials by examining the drug targets and data present for each drug's ability to regulate the aforementioned three hallmark pathways in AAA progression. Historically, drugs that were examined in interventional clinical trials for treatment of AAA were repurposed therapeutics. Novel treatments (biologics, small-molecule compounds etc.) have not been able to reach the clinic, stalling out in pre-clinical studies. Here we discuss the backgrounds of previous investigational drugs in hopes of better informing future development of potential therapeutics. Overall, the highlighted themes discussed here stress the importance of both centralized anti-inflammatory drug targets and rigor of translatability. Exceedingly few murine studies have examined an intervention-based drug treatment in halting further growth of an established AAA despite interventional treatment being the therapeutic approach taken to treat AAA in a clinical setting. Additionally, data suggest that a potentially successful drug target may be a central inflammatory biomarker. Specifically, one that can effectively modulate all three hallmark factors of AAA formation, not just inflammation. It is suggested that inhibiting PGE2 formation with an mPGES-1 inhibitor is a leading drug target for AAA treatment to this end.
Collapse
Affiliation(s)
- Lauren M Weaver
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536, USA.
| | - Charles D Loftin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536, USA.
| | - Chang-Guo Zhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536, USA; Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536, USA.
| |
Collapse
|
13
|
Malik MNH, Waqas SFH, Zeitvogel J, Cheng J, Geffers R, Gouda ZAE, Elsaman AM, Radwan AR, Schefzyk M, Braubach P, Auber B, Olmer R, Müsken M, Roesner LM, Gerold G, Schuchardt S, Merkert S, Martin U, Meissner F, Werfel T, Pessler F. Congenital deficiency reveals critical role of ISG15 in skin homeostasis. J Clin Invest 2021; 132:141573. [PMID: 34847081 PMCID: PMC8803340 DOI: 10.1172/jci141573] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/24/2021] [Indexed: 12/02/2022] Open
Abstract
Ulcerating skin lesions are manifestations of human ISG15 deficiency, a type I interferonopathy. However, chronic inflammation may not be their exclusive cause. We describe two siblings with recurrent skin ulcers that healed with scar formation upon corticosteroid treatment. Both had a homozygous nonsense mutation in the ISG15 gene, leading to unstable ISG15 protein lacking the functional domain. We characterized ISG15–/– dermal fibroblasts, HaCaT keratinocytes, and human induced pluripotent stem cell–derived vascular endothelial cells. ISG15-deficient cells exhibited the expected hyperinflammatory phenotype, but also dysregulated expression of molecules critical for connective tissue and epidermis integrity, including reduced collagens and adhesion molecules, but increased matrix metalloproteinases. ISG15–/– fibroblasts exhibited elevated ROS levels and reduced ROS scavenger expression. As opposed to hyperinflammation, defective collagen and integrin synthesis was not rescued by conjugation-deficient ISG15. Cell migration was retarded in ISG15–/– fibroblasts and HaCaT keratinocytes, but normalized under ruxolitinib treatment. Desmosome density was reduced in an ISG15–/– 3D epidermis model. Additionally, there were loose architecture and reduced collagen and desmoglein expression, which could be reversed by treatment with ruxolitinib/doxycycline/TGF-β1. These results reveal critical roles of ISG15 in maintaining cell migration and epidermis and connective tissue homeostasis, whereby the latter likely requires its conjugation to yet unidentified targets.
Collapse
Affiliation(s)
- Muhammad Nasir Hayat Malik
- Biomarkers for Infectious Diseases, Centre for Experimental and Clinical Infection Research, Twincore, Hannover, Germany
| | - Syed F Hassnain Waqas
- Biomarkers for Infectious Diseases, Centre for Experimental and Clinical Infection Research, Twincore, Hannover, Germany
| | - Jana Zeitvogel
- Institute for Dermatology, Allergology and Venerology, Hannover Medical School (MHH), Hannover, Germany
| | - Jingyuan Cheng
- Experimental Systems Immunology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Robert Geffers
- Genome Analytics, Helmholtz Center for Infection Research, Braunschweig, Germany
| | | | | | - Ahmed R Radwan
- Department of Rheumatology and Rehabilitation, Sohag University, Sohag, Egypt
| | - Matthias Schefzyk
- Institute for Dermatology, Allergology and Venerology, Hannover Medical School (MHH), Hannover, Germany
| | - Peter Braubach
- Institute for Pathology, Hannover Medical School (MHH), Hannover, Germany
| | - Bernd Auber
- Institute for Human Genetics, Hannover Medical School (MHH), Hannover, Germany
| | - Ruth Olmer
- LEBAO, Hannover Medical School (MHH), Hannover, Germany
| | - Mathias Müsken
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lennart M Roesner
- Genome Analytics, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Gisa Gerold
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Sven Schuchardt
- Department of Bio and Environmental Analytics, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | | | - Ulrich Martin
- LEBAO, Hannover Medical School (MHH), Hannover, Germany
| | - Felix Meissner
- Experimental Systems Immunology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Thomas Werfel
- Genome Analytics, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Frank Pessler
- Biomarkers for Infectious Diseases, Centre for Experimental and Clinical Infection Research, Twincore, Hannover, Germany
| |
Collapse
|
14
|
Muhammad W, Zhai Z, Wang S, Gao C. Inflammation-modulating nanoparticles for pneumonia therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1763. [PMID: 34713969 DOI: 10.1002/wnan.1763] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/23/2022]
Abstract
Pneumonia is a common but serious infectious disease, and is the sixth leading cause for death. The foreign pathogens such as viruses, fungi, and bacteria establish an inflammation response after interaction with lung, leading to the filling of bronchioles and alveoli with fluids. Although the pharmacotherapies have shown their great effectiveness to combat pathogens, advanced methods are under developing to treat complicated cases such as virus-infection and lung inflammation or acute lung injury (ALI). The inflammation modulation nanoparticles (NPs) can effectively suppress immune cells and inhibit inflammatory molecules in the lung site, and thereby alleviate pneumonia and ALI. In this review, the pathological inflammatory microenvironments in pneumonia, which are instructive for the design of biomaterials therapy, are summarized. The focus is then paid to the inflammation-modulating NPs that modulate the inflammatory cells, cytokines and chemokines, and microenvironments of pneumonia for better therapeutic effects. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Respiratory Disease.
Collapse
Affiliation(s)
- Wali Muhammad
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Zihe Zhai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Shuqin Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
15
|
Paghdar S, Khan TM, Patel NP, Chandrasekaran S, De Sousa JFM, Tsouklidis N. Doxycycline Therapy for Abdominal Aortic Aneurysm: Inhibitory Effect on Matrix Metalloproteinases. Cureus 2021; 13:e14966. [PMID: 34123662 PMCID: PMC8191685 DOI: 10.7759/cureus.14966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening condition associated with smoking, aging, atherosclerosis, and destruction of the connective tissue in the abdominal aortic wall. Disturbances in the synthesis and degradation of matrix metalloproteinase (MMP) have been known to contribute to the development of AAAs. The only available treatment of AAA is surgical repair. Doxycycline, a tetracycline analog, is thought to have an inhibitory effect on MMPs. Knowing the effect of doxycycline, there may be some favorable effects of the drug to reduce the growth of small AAAs and avoid the need for invasive treatment. This article aims to determine the relationship between doxycycline and the MMPs to prevent the growth of small AAAs. We conducted our review using online resources such as PubMed, Google Scholar, The Journal of Vascular Surgery, and ResearchGate. The result of our study supports the effect of doxycycline in preventing the growth of small AAAs. We conclude that therapeutic treatment with doxycycline in patients with small AAAs can prevent the growth of aneurysms, life-threatening aneurysm rupture, and reduce the need for expensive, invasive treatment.
Collapse
Affiliation(s)
- Smit Paghdar
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Internal Medicine, Surat Municipal Institute of Medical Education and Research (SMIMER), Surat, IND
| | - Taheseen M Khan
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Nishant P Patel
- Internal Medicine, Government Medical College, Surat, Surat, IND.,Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Savitri Chandrasekaran
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Joaquim Francisco Maria De Sousa
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Surgery, S.S. Institute of Medical Sciences and Research Centre, Davangere, IND.,Emergency Medicine, Healthway Hospital, Panaji, IND
| | - Nicholas Tsouklidis
- Health Care Administration, University of Cincinnati Health, Cincinnati, USA.,Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Medicine, Atlantic University School of Medicine, Gros Islet, LCA
| |
Collapse
|
16
|
Sarker H, Haimour A, Toor R, Fernandez-Patron C. The Emerging Role of Epigenetic Mechanisms in the Causation of Aberrant MMP Activity during Human Pathologies and the Use of Medicinal Drugs. Biomolecules 2021; 11:biom11040578. [PMID: 33920915 PMCID: PMC8071227 DOI: 10.3390/biom11040578] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 12/15/2022] Open
Abstract
Matrix metalloproteinases (MMPs) cleave extracellular matrix proteins, growth factors, cytokines, and receptors to influence organ development, architecture, function, and the systemic and cell-specific responses to diseases and pharmacological drugs. Conversely, many diseases (such as atherosclerosis, arthritis, bacterial infections (tuberculosis), viral infections (COVID-19), and cancer), cholesterol-lowering drugs (such as statins), and tetracycline-class antibiotics (such as doxycycline) alter MMP activity through transcriptional, translational, and post-translational mechanisms. In this review, we summarize evidence that the aforementioned diseases and drugs exert significant epigenetic pressure on genes encoding MMPs, tissue inhibitors of MMPs, and factors that transcriptionally regulate the expression of MMPs. Our understanding of human pathologies associated with alterations in the proteolytic activity of MMPs must consider that these pathologies and their medicinal treatments may impose epigenetic pressure on the expression of MMP genes. Whether the epigenetic mechanisms affecting the activity of MMPs can be therapeutically targeted warrants further research.
Collapse
|
17
|
Stepwise candidate drug screening for myopia control by using zebrafish, mouse, and Golden Syrian Hamster myopia models. EBioMedicine 2021; 65:103263. [PMID: 33691248 PMCID: PMC7941086 DOI: 10.1016/j.ebiom.2021.103263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 11/15/2020] [Accepted: 02/11/2021] [Indexed: 12/12/2022] Open
Abstract
Background We developed a preclinical protocol for the screening of candidate drugs able to control myopia and prevent its progression. The protocol uses zebrafish, C57BL/6 mice, and golden Syrian hamster models of myopia. Methods A morpholino (MO) targeting the zebrafish lumican gene (zlum) was injected into single-cell zebrafish embryos, causing excessive expansion of the sclera. A library of 640 compounds with 2 matrix metalloproteinase (MMP) inhibitors (marimastat and batimastat), which have the potential to modulate scleral remodelling, was screened to identify candidates for mitigating scleral diameter expansion in zlum-MO-injected embryos. The myopia-prevention ability of compounds discovered to have superior potency to inhibit scleral expansion was validated over 4 weeks in 4-week-old C57BL/6 mice and 3-week-old golden Syrian hamsters with form-deprivation myopia (FDM). Changes in the refractive error and axial length were investigated. Scleral thickness, morphology of collagen fibrils in the posterior sclera, messenger RNA (mRNA) expressions, and protein levels of transforming growth factor-β2 (TGF-β2), tissue inhibitor of metalloproteinase-2 (TIMP-2), MMP-2, MMP-7, MMP-9, and collagen, type I, alpha 1 (collagen Iα1) were investigated in C57BL/6 mice, and MMP-2, MMP-9, and MMP activity assays were conducted in these mice. Findings In the zebrafish experiment, atropine, marimastat, batimastat, doxycycline, and minocycline were the drugs that most effectively reduced expansion of scleral equatorial diameter. After 28-day treatment in diffuser-wearing mice and 21-day treatment in lid-sutured hamsters, myopic shift and axial elongation were significantly mitigated by eye drops containing 1% atropine, 50 µM marimastat, 5 µM batimastat, or 200 µM doxycycline. MMP-2 mRNA expression in mouse sclera was lower after treatment with atropine, marimastat, batimastat, or doxycycline. The protein levels and activity of MMP-2 and MMP-7 were significantly reduced after treatment with atropine, marimastat, batimastat, doxycycline, and minocycline. Furthermore, scleral thickness and collagen fibril diameter were not lower after treatment with atropine, marimastat, batimastat, or doxycycline than those of occluded eyes. Interpretation Stepwise drug screening in a range of models from zlum-MO-injected zebrafish to rodent FDM models identified effective compounds for preclinical myopia control or prevention. On the basis of the 640 compounds that were screened, MMP inhibitors may offer alternatives for clinical trials. Funding This research was supported by grants from Taiwan's Ministry of Science and Technology and Ministry of Health and Welfare.
Collapse
|
18
|
Singh H, Chauhan P, Singh J, Saurabh S, Gautam CS, Kakkar AK. Concomitant use of dexamethasone and tetracyclines: a potential therapeutic option for the management of severe COVID-19 infection? Expert Rev Clin Pharmacol 2021; 14:315-322. [PMID: 33586566 PMCID: PMC7938652 DOI: 10.1080/17512433.2021.1888714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 02/08/2021] [Indexed: 02/07/2023]
Abstract
Introduction: The global coronavirus disease-2019 (COVID-19) pandemic has posed a critical challenge to the research community as well as to the healthcare systems. Severe COVID-19 patients are at a higher risk of developing serious complications and mortality. There is a dire need for safe and effective pharmacotherapy for addressing unmet needs of these patients. Concomitant use of dexamethasone and tetracyclines, by virtue of their immunomodulatory and other relevant pharmacological properties, offers a potential strategy for synergy aimed at improving clinical outcomes.Areas covered: Here we review the potential benefits of combining dexamethasone and tetracyclines (minocycline or doxycycline) for the management of severe COVID-19 patients. We have critically examined the evidence obtained from in silico, experimental, and clinical research. We have also discussed the plausible mechanisms, advantages, and drawbacks of this proposed combination therapy for managing severe COVID-19.Expert opinion: The concomitant use of dexamethasone and one of the tetracyclines among severe COVID-19 patients offers several advantages in terms of additive immunomodulatory effects, cost-effectiveness, wide-availability, and well-known pharmacological properties including adverse-effect profile and contraindications. There is an urgent need to facilitate pilot studies followed by well-designed and adequately-powered multicentric clinical trials to generate conclusive evidence related to utility of this approach.
Collapse
Affiliation(s)
- Harmanjit Singh
- Department of Pharmacology, Government Medical College and Hospital, Chandigarh, India
| | - Prerna Chauhan
- Multidisciplinary Research Unit, Government Medical College and Hospital, Chandigarh, India
| | - Jasbir Singh
- Department of Pharmacology, Government Medical College and Hospital, Chandigarh, India
- Department of Pharmacology, Rajindra Hospital, Patiala, India
| | - Saurabh Saurabh
- Department of Neurosurgery, Dayanand Medical College and Hospital, Ludhiana, India
| | - CS Gautam
- Department of Pharmacology, Government Medical College and Hospital, Chandigarh, India
| | - Ashish Kumar Kakkar
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
19
|
Dalhoff A. Selective toxicity of antibacterial agents-still a valid concept or do we miss chances and ignore risks? Infection 2021; 49:29-56. [PMID: 33367978 PMCID: PMC7851017 DOI: 10.1007/s15010-020-01536-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/04/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Selective toxicity antibacteribiotics is considered to be due to interactions with targets either being unique to bacteria or being characterized by a dichotomy between pro- and eukaryotic pathways with high affinities of agents to bacterial- rather than eukaryotic targets. However, the theory of selective toxicity oversimplifies the complex modes of action of antibiotics in pro- and eukaryotes. METHODS AND OBJECTIVE This review summarizes data describing multiple modes of action of antibiotics in eukaryotes. RESULTS Aminoglycosides, macrolides, oxazolidinones, chloramphenicol, clindamycin, tetracyclines, glycylcyclines, fluoroquinolones, rifampicin, bedaquillin, ß-lactams inhibited mitochondrial translation either due to binding to mitosomes, inhibition of mitochondrial RNA-polymerase-, topoisomerase 2ß-, ATP-synthesis, transporter activities. Oxazolidinones, tetracyclines, vancomycin, ß-lactams, bacitracin, isoniazid, nitroxoline inhibited matrix-metalloproteinases (MMP) due to chelation with zinc and calcium, whereas fluoroquinols fluoroquinolones and chloramphenicol chelated with these cations, too, but increased MMP activities. MMP-inhibition supported clinical efficacies of ß-lactams and daptomycin in skin-infections, and of macrolides, tetracyclines in respiratory-diseases. Chelation may have contributed to neuroprotection by ß-lactams and fluoroquinolones. Aminoglycosides, macrolides, chloramphenicol, oxazolidins oxazolidinones, tetracyclines caused read-through of premature stop codons. Several additional targets for antibiotics in human cells have been identified like interaction of fluoroquinolones with DNA damage repair in eukaryotes, or inhibition of mucin overproduction by oxazolidinones. CONCLUSION The effects of antibiotics on eukaryotes are due to identical mechanisms as their antibacterial activities because of structural and functional homologies of pro- and eukaryotic targets, so that the effects of antibiotics on mammals are integral parts of their overall mechanisms of action.
Collapse
Affiliation(s)
- Axel Dalhoff
- Christian-Albrechts-University of Kiel, Institue for Infection Medicine, Brunswiker Str. 4, D-24105, Kiel, Germany.
| |
Collapse
|
20
|
Shi X, He L, Zhang SM, Luo J. Human iPS Cell-derived Tissue Engineered Vascular Graft: Recent Advances and Future Directions. Stem Cell Rev Rep 2020; 17:862-877. [PMID: 33230612 DOI: 10.1007/s12015-020-10091-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2020] [Indexed: 12/19/2022]
Abstract
Tissue engineered vascular grafts (TEVGs) generated from human primary cells represent a promising vascular interventional therapy. However, generation and application of these TEVGs may be significantly hindered by the limited accessibility, finite expandability, donor-donor functional variation and immune-incompatibility of primary seed cells from donors. Alternatively, human induced pluripotent stem cells (hiPSCs) offer an infinite source to obtain functional vascular cells in large quantity and comparable quality for TEVG construction. To date, TEVGs (hiPSC-TEVGs) with significant mechanical strength and implantability have been generated using hiPSC-derived seed cells. Despite being in its incipient stage, this emerging field of hiPSC-TEVG research has achieved significant progress and presented promising future potential. Meanwhile, a series of challenges pertaining hiPSC differentiation, vascular tissue engineering technologies and future production and application await to be addressed. Herein, we have composed this review to introduce progress in TEVG generation using hiPSCs, summarize the current major challenges, and encapsulate the future directions of research on hiPSC-based TEVGs. Graphical abstract.
Collapse
Affiliation(s)
- Xiangyu Shi
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.,Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine , Yale School of Medicine, 300 George Street, Room 752, New Haven, CT, 06511, USA
| | - Lile He
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Shang-Min Zhang
- Department of Pathology, Yale School of Medicine, 06520, New Haven, CT, USA
| | - Jiesi Luo
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine , Yale School of Medicine, 300 George Street, Room 752, New Haven, CT, 06511, USA. .,Yale Stem Cell Center, 06520, New Haven, CT, USA.
| |
Collapse
|
21
|
Hosseini V, Mallone A, Mirkhani N, Noir J, Salek M, Pasqualini FS, Schuerle S, Khademhosseini A, Hoerstrup SP, Vogel V. A Pulsatile Flow System to Engineer Aneurysm and Atherosclerosis Mimetic Extracellular Matrix. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000173. [PMID: 32596117 PMCID: PMC7312268 DOI: 10.1002/advs.202000173] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Indexed: 06/11/2023]
Abstract
Alterations of blood flow patterns strongly correlate with arterial wall diseases such as atherosclerosis and aneurysm. Here, a simple, pumpless, close-loop, easy-to-replicate, and miniaturized flow device is introduced to concurrently expose 3D engineered vascular smooth muscle tissues to high-velocity pulsatile flow versus low-velocity disturbed flow conditions. Two flow regimes are distinguished, one that promotes elastin and impairs collagen I assembly, while the other impairs elastin and promotes collagen assembly. This latter extracellular matrix (ECM) composition shares characteristics with aneurysmal or atherosclerotic tissue phenotypes, thus recapitulating crucial hallmarks of flow-induced tissue morphogenesis in vessel walls. It is shown that the mRNA levels of ECM of collagens and elastin are not affected by the differential flow conditions. Instead, the differential gene expression of matrix metalloproteinase (MMP) and their inhibitors (TIMPs) is flow-dependent, and thus drives the alterations in ECM composition. In further support, treatment with doxycycline, an MMP inhibitor and a clinically used drug to treat vascular diseases, halts the effect of low-velocity flow on the ECM remodeling. This illustrates how the platform can be exploited for drug efficacy studies by providing crucial mechanistic insights into how different therapeutic interventions may affect tissue growth and ECM assembly.
Collapse
Affiliation(s)
- Vahid Hosseini
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH ZurichZurich8093Switzerland
- Present address:
Department of BioengineeringUniversity of California‐Los AngelesLos AngelesCA90095USA
| | - Anna Mallone
- Institute for Regenerative Medicine (IREM)University of Zurich and Wyss Translational Center ZurichZurich8952Switzerland
| | - Nima Mirkhani
- Responsive Biomedical Systems LabInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH ZurichZurich8093Switzerland
| | - Jerome Noir
- Institute of GeophysicsDepartment of Earth SciencesETH ZurichZurich8092Switzerland
| | - Mehdi Salek
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyBostonMA02139USA
| | - Francesco Silvio Pasqualini
- Institute for Regenerative Medicine (IREM)University of Zurich and Wyss Translational Center ZurichZurich8952Switzerland
- Synthetic Physiology LaboratoryDepartment of Civil Engineering and ArchitectureUniversity of PaviaPavia27100Italy
| | - Simone Schuerle
- Responsive Biomedical Systems LabInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH ZurichZurich8093Switzerland
| | - Ali Khademhosseini
- Department of BioengineeringUniversity of California‐Los AngelesLos AngelesCA90095USA
| | - Simon P. Hoerstrup
- Institute for Regenerative Medicine (IREM)University of Zurich and Wyss Translational Center ZurichZurich8952Switzerland
| | - Viola Vogel
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH ZurichZurich8093Switzerland
| |
Collapse
|
22
|
Dean RS, Kahat DH, Graden NR, DePhillipo NN, LaPrade RF. Doxycycline improves tendon and cartilage pathologies in preclinical studies: current concepts. J ISAKOS 2020. [DOI: 10.1136/jisakos-2020-000451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
23
|
Patlolla VGR, Peter Holbrook W, Gizurarson S, Kristmundsdottir T. Doxycycline and Monocaprin In Situ Hydrogel: Effect on Stability, Mucoadhesion and Texture Analysis and In Vitro Release. Gels 2019; 5:E47. [PMID: 31835322 PMCID: PMC6956273 DOI: 10.3390/gels5040047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 11/17/2022] Open
Abstract
The aim of this study was to develop a stable aqueous formulation containing a combination of doxycycline and monocaprin in clinically relevant concentrations. Increase in expression of Matrix metalloproteinases (MMPs) and microbial role in oral diseases is well established and the combination of above active ingredients could be potentially beneficial in treatment of oral mucosal conditions. The hydrogels containing different concentrations of doxycycline and monocaprin in the presence and absence of stabilizing excipients were developed and their stabilities were studied at 4 °C for up to 1 year. The drug-drug interaction was evaluated using Fourier-transform infrared spectroscopy (FTIR). The addition of monocaprin on doxycycline in situ hydrogel's mucoadhesiveness, texture properties and drug release mechanism was studied. The addition of monocaprin negatively affected the doxycycline stability and was concentration dependent, whereas monocaprin was stable up to 1 year. Doxycycline did not interfere with the anti-Candidal activity of monocaprin. Furthermore, the presence of monocaprin significantly affected the formulation hardness, compressibility and adhesiveness. Monocaprin and doxycycline release followed zero order kinetics and the release mechanism was, by anomalous (non-Fickian) diffusion. The addition of monocaprin increased the drug release time and altered the release mechanism. It is possible to stabilize doxycycline in the presence of monocaprin up to 1 year at 4 °C.
Collapse
Affiliation(s)
- Venu Gopal Reddy Patlolla
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, 107 Reykjavik, Iceland; (V.G.R.P.); (S.G.)
- Faculty of Odontology, University of Iceland, Vatnsmýrarveg 16, 101 Reykjavík, Iceland;
| | | | - Sveinbjorn Gizurarson
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, 107 Reykjavik, Iceland; (V.G.R.P.); (S.G.)
| | - Thordis Kristmundsdottir
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, 107 Reykjavik, Iceland; (V.G.R.P.); (S.G.)
| |
Collapse
|
24
|
Clemens DL, Duryee MJ, Hall JH, Thiele GM, Mikuls TR, Klassen LW, Zimmerman MC, Anderson DR. Relevance of the antioxidant properties of methotrexate and doxycycline to their treatment of cardiovascular disease. Pharmacol Ther 2019; 205:107413. [PMID: 31626869 DOI: 10.1016/j.pharmthera.2019.107413] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/15/2019] [Indexed: 12/21/2022]
Abstract
Many medications exhibit clinical benefits that are unrelated to their primary therapeutic uses. In many cases, the mechanisms underpinning these pleotropic effects are unknown. Two commonly prescribed medications that exhibit pleotropic benefits in cardiovascular disease and other diseases associated with chronic inflammation are methotrexate (MTX) and doxycycline (DOX). The vast majority of cardiovascular disease is associated with atherosclerosis. Because atherosclerosis is a chronic inflammatory disease, possible mechanisms by which MTX and DOX reduce inflammation have been investigated. Interestingly, the primary structure of both of these medications contain aromatic phenolic rings, which resemble polyphenols that are known to possess antioxidant activity. Inflammation and oxidative stress are intimately related. Inflammation promotes oxidative stress, which in turn leads to further inflammation; in this way, oxidative stress and inflammation can establish a self-perpetuating cycle. It has been shown that MTX and DOX act as antioxidants and are capable of scavenging free radicals and the reactive oxygen species (ROS) superoxide (O2-). Furthermore, both MTX and DOX inhibit the formation of malondialdehyde acetaldehyde (MAA) adducts, products of oxidative stress and lipid peroxidation. Importantly, MAA-adducts are highly immunogenic and initiate inflammatory responses; thereby, fueling the cycle of inflammation and oxidative stress that results in chronic inflammation. Thus, reducing the formation of MAA-adducts may ameliorate inflammation that leads to ROS production and in this way, break the self-sustaining cycle of oxidative stress and inflammation. It is possible that the under-recognized antioxidant properties of these medications may be a mechanism by which they and other medications provide pleotropic benefit in the treatment of chronic inflammatory disease.
Collapse
Affiliation(s)
- Dahn L Clemens
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE, 68198-2265, United States; Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave., Omaha, NE, 68105, United States; Fred and Pamela Buffet Cancer Center, Nebraska Medical Center, Omaha, NE, 68114, United States
| | - Michael J Duryee
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE, 68198-2265, United States; Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave., Omaha, NE, 68105, United States
| | - Johnathan H Hall
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE, 68198-2265, United States
| | - Geoffrey M Thiele
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE, 68198-2265, United States; Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave., Omaha, NE, 68105, United States
| | - Ted R Mikuls
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE, 68198-2265, United States; Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave., Omaha, NE, 68105, United States
| | - Lynell W Klassen
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE, 68198-2265, United States
| | - Matthew C Zimmerman
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE, 68198-2265, United States
| | - Daniel R Anderson
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE, 68198-2265, United States.
| |
Collapse
|
25
|
Doxycycline inhibits electric field-induced migration of non-small cell lung cancer (NSCLC) cells. Sci Rep 2019; 9:8094. [PMID: 31147570 PMCID: PMC6542854 DOI: 10.1038/s41598-019-44505-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 05/15/2019] [Indexed: 01/01/2023] Open
Abstract
Adenocarcinoma, large cell carcinoma and squamous cell carcinoma are the most commonly diagnosed subtypes of non-small cell lung cancers (NSCLC). Numerous lung cancer cell types have exhibited electrotaxis under direct current electric fields (dcEF). Physiological electric fields (EF) play key roles in cancer cell migration. In this study, we investigated electrotaxis of NSCLC cells, including human large cell lung carcinoma NCI-H460 and human lung squamous cell carcinoma NCI-H520 cells. Non-cancerous MRC-5 lung fibroblasts were included as a control. After dcEF stimulation, NCI-H460 and NCI-H520 cells, which both exhibit epithelial-like morphology, migrated towards the cathode, while MRC-5 cells, which have fibroblast-like morphology, migrated towards the anode. The effect of doxycycline, a common antibiotic, on electrotaxis of MRC-5, NCI-H460 and NCI-H520 cells was examined. Doxycycline enhanced the tested cells’ motility but inhibited electrotaxis in the NSCLC cells without inhibiting non-cancerous MRC-5 cells. Based on our finding, further in-vivo studies could be devised to investigate the metastasis inhibition effect of doxycycline in an organism level.
Collapse
|
26
|
Samartzis EP, Fink D, Stucki M, Imesch P. Doxycycline reduces MMP-2 activity and inhibits invasion of 12Z epithelial endometriotic cells as well as MMP-2 and -9 activity in primary endometriotic stromal cells in vitro. Reprod Biol Endocrinol 2019; 17:38. [PMID: 30981279 PMCID: PMC6462312 DOI: 10.1186/s12958-019-0481-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 04/04/2019] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Matrix metalloproteinases (MMPs), especially the gelatinases MMP-2 and MMP-9, play a crucial role in the pathogenesis of endometriosis by enabling invasion. Doxycycline is a well-tolerated antibiotic and a potent MMP-inhibitor in subantimicrobial doses. METHODS Gelatin zymography and activity assays were used to detect latent and active MMP-2 and -9 in cell culture supernatants of immortalized epithelial (12Z) and two isolates of primary endometriotic stromal cells treated with doxycycline. The invasiveness of 12Z endometriotic cells treated with doxycycline was assessed in matrigel-coated invasion chambers. The effect on latent and active MMP-2 expression of the combination of progesterone and doxycycline was tested in 12Z. RESULTS Doxycycline significantly reduced the MMP-2 activity and pro-MMP-2 expression in 12Z and the MMP-2 and -9 activity as well as expression of pro-MMP-2 and -9 in primary endometriotic stromal cells. The percentage of 12Z cells invading through a matrigel-coated membrane was reduced to 65 and 22% of the control after treatment with doxycycline at doses of 1 μg/ml and 10 μg/ml, respectively. Furthermore, a combination of progesterone and doxycycline showed an additive effect in low doses on the reduction of MMP-2 activity and pro-MMP2 expression in 12Z endometriotic cells. CONCLUSIONS In conclusion, the MMP-inhibiting features of subantimicrobial-dose doxycycline may be further evaluated as a well-tolerable additional therapeutic approach, e.g. in combination with progestins such as dienogest, in patients with infiltrative endometriosis with insufficient response to current medical treatment options.
Collapse
Affiliation(s)
- Eleftherios P. Samartzis
- 0000 0004 0478 9977grid.412004.3Division of Gynecology, University Hospital Zurich, Frauenklinikstrasse 10, CH-8091 Zürich, Switzerland
| | - Daniel Fink
- 0000 0004 0478 9977grid.412004.3Division of Gynecology, University Hospital Zurich, Frauenklinikstrasse 10, CH-8091 Zürich, Switzerland
| | - Manuel Stucki
- 0000 0004 0478 9977grid.412004.3Division of Gynecology, University Hospital Zurich, Frauenklinikstrasse 10, CH-8091 Zürich, Switzerland
| | - Patrick Imesch
- 0000 0004 0478 9977grid.412004.3Division of Gynecology, University Hospital Zurich, Frauenklinikstrasse 10, CH-8091 Zürich, Switzerland
| |
Collapse
|
27
|
Cui JZ, Lee L, Sheng X, Chu F, Gibson CP, Aydinian T, Walker DC, Sandor GGS, Bernatchez P, Tibbits GF, van Breemen C, Esfandiarei M. In vivo characterization of doxycycline-mediated protection of aortic function and structure in a mouse model of Marfan syndrome-associated aortic aneurysm. Sci Rep 2019; 9:2071. [PMID: 30765726 PMCID: PMC6376062 DOI: 10.1038/s41598-018-38235-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 12/21/2018] [Indexed: 12/31/2022] Open
Abstract
Aortic aneurysm is the most life-threatening complication in Marfan syndrome (MFS) patients. Doxycycline, a nonselective matrix metalloproteinases inhibitor, was reported to improve the contractile function and elastic fiber structure and organization in a Marfan mouse aorta using ex vivo small chamber myography. In this study, we assessed the hypothesis that a long-term treatment with doxycycline would reduce aortic root growth, improve aortic wall elasticity as measured by pulse wave velocity, and improve the ultrastructure of elastic fiber in the mouse model of MFS. In our study, longitudinal measurements of aortic root diameters using high-resolution ultrasound imaging display significantly decreased aortic root diameters and lower pulse wave velocity in doxycycline-treated Marfan mice starting at 6 months as compared to their non-treated MFS counterparts. In addition, at the ultrastructural level, our data show that long-term doxycycline treatment corrects the irregularities of elastic fibers within the aortic wall of Marfan mice to the levels similar to those observed in control subjects. Our findings underscore the key role of matrix metalloproteinases during the progression of aortic aneurysm, and provide new insights into the potential therapeutic value of doxycycline in blocking MFS-associated aortic aneurysm.
Collapse
Affiliation(s)
- Jason Z Cui
- Department of Anesthesiology, Pharmacology and Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada.,Department of Cardiothoracic Surgery, School of Medicine, Stanford University, Palo Alto, California, USA
| | - Ling Lee
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Xiaoye Sheng
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Fanny Chu
- Department of Anesthesiology, Pharmacology and Therapeutics, Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Christine P Gibson
- Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Glendale, Arizona, USA
| | - Taline Aydinian
- Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Glendale, Arizona, USA
| | - David C Walker
- Department of Anesthesiology, Pharmacology and Therapeutics, Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - George G S Sandor
- Children's Heart Centre, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology and Therapeutics, Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Glen F Tibbits
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Cornelis van Breemen
- Department of Anesthesiology, Pharmacology and Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Mitra Esfandiarei
- Department of Anesthesiology, Pharmacology and Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada. .,Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Glendale, Arizona, USA.
| |
Collapse
|
28
|
Mountain DJH, Kirkpatrick SS, Arnold JD, Buckley MR, McNally MM, Stevens SL, Freeman MB, Grandas OH. The Efficacy of Systemic Doxycycline Administration as an Inhibitor of Intimal Hyperplasia after Balloon Angioplasty Arterial Injury. Ann Vasc Surg 2019; 57:201-209. [PMID: 30684618 DOI: 10.1016/j.avsg.2018.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/25/2018] [Accepted: 11/18/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Intimal hyperplasia (IH) is the most common indicator for secondary intervention in peripheral vascular disease. Matrix metalloproteinases (MMPs) play a role in IH development due to their degradation of the extracellular matrix. Doxycycline (Doxy), a member of the tetracycline family of antibiotics, is a potent MMP inhibitor. We have previously shown that Doxy inhibits MMP activity and vascular smooth muscle cell migration in vitro. We hypothesized that Doxy would decrease MMP activity in vivo and inhibit the development of IH in a rodent model of vascular injury. METHODS AND RESULTS Doxy (400 mg/pellet) was delivered by a slow-release pellet implanted 3 days prior to or at the time of balloon angioplasty (BA) of the common carotid artery in female rats. At 14 days post-BA, intima-to-media (I:M) ratios were 0.77 ± 0.21 and 1.04 ± 0.32 in the Doxy treated groups, respectively, compared to 1.25 ± 0.26 in the control group (P = not significant; n = 3). Additionally, the tested dose of Doxy in either group had no inhibitory effect on membrane type 1-MMP or MMP-2 tissue levels, as measured by immunohistochemistry, or on systemic levels of MMP, as measured by total MMP serum levels using enzyme-linked immunosorbent assay. At 14 days post-BA, VSMC proliferation in the injured artery was increased to Doxy treatment prior to and at the time of surgery (23.5 ± 3.4 and 27.2 ± 3.9%, respectively), compared to control (11.4 ± 0.4%; n = 3), as measured by proliferating cellular nuclear antigen immunostaining. CONCLUSIONS In our in vivo model of vascular injury, systemic Doxy administration prior to or at the time of vascular injury does not significantly hinder the progression of IH development. Additional doses and routes of administration could be examined in order to correlate therapeutic serum levels of Doxy with effective MMP inhibition in serum and arterial tissue. However, alternative drug delivery systems are needed in order to optimize therapeutic administration of targeted MMP inhibitors for the prevention of IH development.
Collapse
Affiliation(s)
- Deidra J H Mountain
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, TN
| | - Stacy S Kirkpatrick
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, TN
| | - Joshua D Arnold
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, TN
| | - M Ryan Buckley
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, TN
| | - Michael M McNally
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, TN
| | - Scott L Stevens
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, TN
| | - Michael B Freeman
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, TN
| | - Oscar H Grandas
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, TN.
| |
Collapse
|
29
|
Clemens DL, Duryee MJ, Sarmiento C, Chiou A, McGowan JD, Hunter CD, Schlichte SL, Tian J, Klassen LW, O'Dell JR, Thiele GM, Mikuls TR, Zimmerman MC, Anderson DR. Novel Antioxidant Properties of Doxycycline. Int J Mol Sci 2018; 19:ijms19124078. [PMID: 30562944 PMCID: PMC6321135 DOI: 10.3390/ijms19124078] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/13/2018] [Accepted: 12/13/2018] [Indexed: 12/12/2022] Open
Abstract
Doxycycline (DOX), a derivative of tetracycline, is a broad-spectrum antibiotic that exhibits a number of therapeutic activities in addition to its antibacterial properties. For example, DOX has been used in the management of a number of diseases characterized by chronic inflammation. One potential mechanism by which DOX inhibits the progression of these diseases is by reducing oxidative stress, thereby inhibiting subsequent lipid peroxidation and inflammatory responses. Herein, we tested the hypothesis that DOX directly scavenges reactive oxygen species (ROS) and inhibits the formation of redox-mediated malondialdehyde-acetaldehyde (MAA) protein adducts. Using a cell-free system, we demonstrated that DOX scavenged reactive oxygen species (ROS) produced during the formation of MAA-adducts and inhibits the formation of MAA-protein adducts. To determine whether DOX scavenges specific ROS, we examined the ability of DOX to directly scavenge superoxide and hydrogen peroxide. Using electron paramagnetic resonance (EPR) spectroscopy, we found that DOX directly scavenged superoxide, but not hydrogen peroxide. Additionally, we found that DOX inhibits MAA-induced activation of Nrf2, a redox-sensitive transcription factor. Together, these findings demonstrate the under-recognized direct antioxidant property of DOX that may help to explain its therapeutic potential in the treatment of conditions characterized by chronic inflammation and increased oxidative stress.
Collapse
Affiliation(s)
- Dahn L Clemens
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, USA.
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, USA.
- Fred and Pamela Buffet Cancer Center, Omaha, NE 68114, USA.
| | - Michael J Duryee
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, USA.
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, USA.
| | - Cleofes Sarmiento
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, USA.
| | - Andrew Chiou
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, USA.
| | - Jacob D McGowan
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, USA.
| | - Carlos D Hunter
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, USA.
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, USA.
| | - Sarah L Schlichte
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, USA.
| | - Jun Tian
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, USA.
| | - Lynell W Klassen
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, USA.
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, USA.
| | - James R O'Dell
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, USA.
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, USA.
| | - Geoffrey M Thiele
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, USA.
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, USA.
| | - Ted R Mikuls
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, USA.
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, USA.
| | - Matthew C Zimmerman
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, USA.
| | - Daniel R Anderson
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, USA.
| |
Collapse
|
30
|
Kumar G, Patnaik R. Inhibition of Gelatinases (MMP-2 and MMP-9) by Withania somnifera Phytochemicals Confers Neuroprotection in Stroke: An In Silico Analysis. Interdiscip Sci 2018; 10:722-733. [PMID: 28488219 DOI: 10.1007/s12539-017-0231-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 01/25/2017] [Accepted: 04/01/2017] [Indexed: 01/24/2023]
Abstract
A stroke or cerebrovascular accident is a serious, life-threatening medical condition that occurs when the blood supply to part of the brain is severely reduced or cut off, depriving brain tissue of oxygen and nutrients. Studies suggested that level of gelatinases (MMP-2 and MMP-9) usually increases in the brain after stroke. The elevated activity of gelatinases plays the deleterious role in ischemic stroke, hemorrhagic stroke and perinatal hypoxic-ischemic brain injury. Therefore, matrix metalloproteinase (MMP)-2 and MMP-9 inhibition have therapeutic importance in stroke condition. Present in silico study investigates whether Withania somnifera (WS) phytochemicals inhibit the MMP-2 and MMP-9 by binding to the catalytic domain, as similar to their inhibitor or not. For that, we performed molecular docking study to evaluate the gelatinases-inhibitory potential of 36 WS phytochemicals, which compared with gelatinases inhibitors viz. hydroxamic acid, quercetin, doxycycline, minocycline and reverse hydroxamate. The results suggest that 28 out of 36 WS phytochemicals show higher affinity for MMP-2 owing to bind with active site residues of S1'-pocket with lower binding energy and smaller inhibition constant (Ki) than considered inhibitors. As well as, withanolide G and withafastuosin E show higher affinity for MMP-9 than reverse hydroxamate inhibitor. These phytochemicals have neuroprotective potential as an inherently useful oral drug to combat ischemic and hemorrhagic stroke mediated by gelatinases.
Collapse
Affiliation(s)
- Gaurav Kumar
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, UP, 221005, India.
| | - Ranjana Patnaik
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, UP, 221005, India
| |
Collapse
|
31
|
Cook R, Sarker H, Fernandez-Patron C. Pathologies of matrix metalloproteinase-2 underactivity: a perspective on a neglected condition 1. Can J Physiol Pharmacol 2018; 97:486-492. [PMID: 30457883 DOI: 10.1139/cjpp-2018-0525] [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] [Indexed: 12/17/2022]
Abstract
A member of the matrix metalloproteinase family, matrix metalloproteinase-2 (MMP-2, gelatinase A), has been extensively studied for its role in both normal physiology and pathological processes. Whereas most research efforts in recent years have investigated the pathologies associated with MMP-2 overactivity, the pathological mechanisms elicited by MMP-2 underactivity are less well understood. Here, we distinguish between 2 states and describe their causes: (i) MMP-2 deficiency (complete loss of MMP-2 activity) and (ii) MMP-2 insufficiency (defined as MMP-2 activity below baseline levels). Further, we review the biology of MMP-2, summarizing the current literature on MMP-2 underactivity in both mice and humans, and describe research being conducted by our lab towards improving our understanding of the pathological mechanisms elicited by MMP-2 deficiency/insufficiency. We think that this research could stimulate the discovery of new therapeutic approaches for managing pathologies associated with MMP-2 underactivity. Moreover, similar concepts could apply to other members of the matrix metalloproteinase family.
Collapse
Affiliation(s)
- Ryan Cook
- a Department of Biochemistry, Faculty of Medicine and Dentistry, 3-19 Medical Sciences Building, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Hassan Sarker
- a Department of Biochemistry, Faculty of Medicine and Dentistry, 3-19 Medical Sciences Building, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Carlos Fernandez-Patron
- b Department of Biochemistry, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, 3-19 Medical Sciences Building, University of Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
32
|
Galior K, Ma VPY, Liu Y, Su H, Baker N, Panettieri RA, Wongtrakool C, Salaita K. Molecular Tension Probes to Investigate the Mechanopharmacology of Single Cells: A Step toward Personalized Mechanomedicine. Adv Healthc Mater 2018; 7:e1800069. [PMID: 29785773 PMCID: PMC6105437 DOI: 10.1002/adhm.201800069] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/15/2018] [Indexed: 01/03/2023]
Abstract
Given that dysregulation of mechanics contributes to diseases ranging from cancer metastasis to lung disease, it is important to develop methods for screening the efficacy of drugs that target cellular forces. Here, nanoparticle-based tension sensors are used to quantify the mechanical response of individual cells upon drug treatment. As a proof-of-concept, the activity of bronchodilators is tested on human airway smooth muscle cells derived from seven donors, four of which are asthmatic. It is revealed that airway smooth muscle cells isolated from asthmatic donors exhibit greater traction forces compared to the control donors. Additionally, the mechanical signal is abolished using myosin inhibitors or further enhanced in the presence of inflammatory inducers, such as nicotine. Using the signal generated by the probes, single-cell dose-response measurements are performed to determine the "mechano" effective concentration (mechano-EC50 ) of albuterol, a bronchodilator, which reduces integrin forces by 50%. Mechano-EC50 values for each donor present discrete readings that are differentially enhanced as a function of nicotine treatment. Importantly, donor mechano-EC50 values varied by orders of magnitude, suggesting significant variability in their sensitivity to nicotine and albuterol treatment. To the best of the authors' knowledge, this is the first study harnessing a piconewton tension sensor platform for mechanopharmacology.
Collapse
Affiliation(s)
- Kornelia Galior
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | | | - Yang Liu
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Hanquan Su
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Nusaiba Baker
- Emory University School of Medicine, Emory University, Atlanta, GA, 30307, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Cherry Wongtrakool
- Emory University School of Medicine, Emory University, Atlanta, GA, 30307, USA
- Atlanta Veterans Affairs Medical Center, Decatur, GA, 30033, USA
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| |
Collapse
|
33
|
Giraud A, Zeboudj L, Vandestienne M, Joffre J, Esposito B, Potteaux S, Vilar J, Cabuzu D, Kluwe J, Seguier S, Tedgui A, Mallat Z, Lafont A, Ait-Oufella H. Gingival fibroblasts protect against experimental abdominal aortic aneurysm development and rupture through tissue inhibitor of metalloproteinase-1 production. Cardiovasc Res 2018; 113:1364-1375. [PMID: 28582477 DOI: 10.1093/cvr/cvx110] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/31/2017] [Indexed: 11/14/2022] Open
Abstract
Aims Abdominal aortic aneurysm (AAA), frequently diagnosed in old patients, is characterized by chronic inflammation, vascular cell apoptosis and metalloproteinase-mediated extracellular matrix destruction. Despite improvement in the understanding of the pathophysiology of aortic aneurysm, no pharmacological treatment is yet available to limit dilatation and/or rupture. We previously reported that human gingival fibroblasts (GFs) can reduce carotid artery dilatation in a rabbit model of elastase-induced aneurysm. Here, we sought to investigate the mechanisms of GF-mediated vascular protection in two different models of aortic aneurysm growth and rupture in mice. Methods and results In vitro, mouse GFs proliferated and produced large amounts of anti-inflammatory cytokines and tissue inhibitor of metalloproteinase-1 (Timp-1). GFs deposited on the adventitia of abdominal aorta survived, proliferated, and organized as a layer structure. Furthermore, GFs locally produced Il-10, TGF-β, and Timp-1. In a mouse elastase-induced AAA model, GFs prevented both macrophage and lymphocyte accumulations, matrix degradation, and aneurysm growth. In an Angiotensin II/anti-TGF-β model of aneurysm rupture, GF cell-based treatment limited the extent of aortic dissection, prevented abdominal aortic rupture, and increased survival. Specific deletion of Timp-1 in GFs abolished the beneficial effect of cell therapy in both AAA mouse models. Conclusions GF cell-based therapy is a promising approach to inhibit aneurysm progression and rupture through local production of Timp-1.
Collapse
Affiliation(s)
- Andreas Giraud
- Inserm U970, Paris Cardiovascular Research Center, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Lynda Zeboudj
- Inserm U970, Paris Cardiovascular Research Center, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marie Vandestienne
- Inserm U970, Paris Cardiovascular Research Center, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jérémie Joffre
- Inserm U970, Paris Cardiovascular Research Center, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bruno Esposito
- Inserm U970, Paris Cardiovascular Research Center, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Stéphane Potteaux
- Inserm U970, Paris Cardiovascular Research Center, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - José Vilar
- Inserm U970, Paris Cardiovascular Research Center, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Daniela Cabuzu
- Inserm U970, Paris Cardiovascular Research Center, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Johannes Kluwe
- Department of Gastroenterology & Hepatology, Hamburg University Medical Center, Hamburg, Germany
| | - Sylvie Seguier
- Inserm U970, Paris Cardiovascular Research Center, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Alain Tedgui
- Inserm U970, Paris Cardiovascular Research Center, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Ziad Mallat
- Inserm U970, Paris Cardiovascular Research Center, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Antoine Lafont
- Inserm U970, Paris Cardiovascular Research Center, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Hafid Ait-Oufella
- Inserm U970, Paris Cardiovascular Research Center, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Medical Intensive Care Unit, Hôpital Saint-Antoine, AP-HP, Université Pierre-et-Marie Curie, Paris, France
| |
Collapse
|
34
|
Nishihama K, Yasuma T, Yano Y, D' Alessandro-Gabazza CN, Toda M, Hinneh JA, Baffour Tonto P, Takeshita A, Totoki T, Mifuji-Moroka R, Kobayashi T, Iwasa M, Takei Y, Morser J, Cann I, Gabazza EC. Anti-apoptotic activity of human matrix metalloproteinase-2 attenuates diabetes mellitus. Metabolism 2018; 82:88-99. [PMID: 29366755 DOI: 10.1016/j.metabol.2018.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/05/2018] [Accepted: 01/18/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Chronic progression of diabetes is associated with decreased pancreatic islet mass due to apoptosis of β-cells. Patients with diabetes have increased circulating matrix metalloproteinase-2 (MMP2); however, the physiological significance has remained elusive. This study tested the hypothesis that MMP2 inhibits cell apoptosis, including islet β-cells. METHODS Samples from diabetic patients and newly developed transgenic mice overexpressing human MMP2 (hMMP2) were harnessed, and diabetes was induced with streptozotocin. RESULTS Circulating hMMP2 was significantly increased in diabetic patients compared to controls and significantly correlated with the serum C-peptide levels. The diabetic hMMP2 transgenic mice showed significant improvements in glycemia, glucose tolerance and insulin secretion compared to diabetic wild type mice. Importantly, the increased hMMP2 levels in mice correlated with significant reduction in islet β-cell apoptosis compared to wild-type counterparts, and an inhibitor of hMMP2 reversed this mitigating activity against diabetes. The increased activation of Akt and BAD induced by hMMP2 in β-cells compared to controls, links this signaling pathway to the anti-apoptotic activity of hMMP2, a property that was reversible by both an hMMP2 inhibitor and antibody against integrin-β3. CONCLUSION Overall, this study demonstrates that increased expression of hMMP2 may attenuate the severity of diabetes by protecting islet β-cells from apoptosis through an integrin-mediated activation of the Akt/BAD pathway.
Collapse
Affiliation(s)
- Kota Nishihama
- Department of Diabetes, Metabolism and Endocrinology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Taro Yasuma
- Department of Diabetes, Metabolism and Endocrinology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan; Department of Immunology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Yutaka Yano
- Department of Diabetes, Metabolism and Endocrinology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Corina N D' Alessandro-Gabazza
- Department of Immunology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan; Microbiome Metabolic Engineering Theme, Carl R. Woese Biology Institute for Genomic Biology, Department of Animal Sciences, Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States
| | - Masaaki Toda
- Department of Immunology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Josephine A Hinneh
- Department of Immunology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Prince Baffour Tonto
- Department of Immunology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Atsuro Takeshita
- Department of Diabetes, Metabolism and Endocrinology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Toshiaki Totoki
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Rumi Mifuji-Moroka
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Tetsu Kobayashi
- Department of Pulmonary and Critical Care Medicine, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Motoh Iwasa
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Yoshiyuki Takei
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - John Morser
- Division of Hematology, Stanford School of Medicine, 269 Campus Drive, CCSR 1155, Stanford, CA 94305-5156, United States
| | - Isaac Cann
- Microbiome Metabolic Engineering Theme, Carl R. Woese Biology Institute for Genomic Biology, Department of Animal Sciences, Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States
| | - Esteban C Gabazza
- Department of Immunology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan.
| |
Collapse
|
35
|
Abstract
INTRODUCTION Lipopolysaccharide (LPS) is known to induce vascular derangements. The pathophysiology involved therein is unknown, but matrix metalloproteinases (MMPs) may be an important mediator. We hypothesized that in vitro LPS provokes vascular permeability, damages endothelial structural proteins, and increases MMP activity; that in vivo LPS increases permeability and fluid requirements; and that the MMP inhibitor doxycycline mitigates such changes. METHODS Rat lung microvascular endothelial cells were divided into four groups: control, LPS, LPS plus doxycycline, and doxycycline. Permeability, structural proteins β-catenin and Filamentous-actin, and MMP-9 activity were examined. Sprauge Dawley rats were divided into sham, IV LPS, and IV LPS plus IV doxycycline groups. Mesenteric postcapillary venules were observed. Blood pressure was measured as animals were resuscitated and fluid requirements were compared. Statistical analysis was conducted using Student's t-test and ANOVA. RESULTS In vitro LPS increased permeability, damaged adherens junctions, induced actin stress fiber formation, and increased MMP-9 enzyme activity. In vivo, IV LPS administration induced vascular permeability. During resuscitation, significantly more fluid was necessary to maintain normotension in the IV LPS group. Doxycycline mitigated all derangements observed. CONCLUSIONS We conclude that LPS increases permeability, damages structural proteins, and increases MMP-9 activity in endothelial cells. Additionally, endotoxemia induces hyperpermeability and increases the amount of IV fluid required to maintain normotension in vivo. Doxycycline mitigates such changes both in vitro and in vivo. Our findings illuminate the possible role of matrix metalloproteinases in the pathophysiology of lipopolysaccharide-induced microvascular hyperpermeability and pave the way for better understanding and treatment of this process.
Collapse
|
36
|
Jankowsky JL, Zheng H. Practical considerations for choosing a mouse model of Alzheimer's disease. Mol Neurodegener 2017; 12:89. [PMID: 29273078 PMCID: PMC5741956 DOI: 10.1186/s13024-017-0231-7] [Citation(s) in RCA: 318] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 12/07/2017] [Indexed: 01/06/2023] Open
Abstract
Alzheimer’s disease (AD) is behaviorally identified by progressive memory impairment and pathologically characterized by the triad of β-amyloid plaques, neurofibrillary tangles, and neurodegeneration. Genetic mutations and risk factors have been identified that are either causal or modify the disease progression. These genetic and pathological features serve as basis for the creation and validation of mouse models of AD. Efforts made in the past quarter-century have produced over 100 genetically engineered mouse lines that recapitulate some aspects of AD clinicopathology. These models have been valuable resources for understanding genetic interactions that contribute to disease and cellular reactions that are engaged in response. Here we focus on mouse models that have been widely used stalwarts of the field or that are recently developed bellwethers of the future. Rather than providing a summary of each model, we endeavor to compare and contrast the genetic approaches employed and to discuss their respective advantages and limitations. We offer a critical account of the variables which may contribute to inconsistent findings and the factors that should be considered when choosing a model and interpreting the results. We hope to present an insightful review of current AD mouse models and to provide a practical guide for selecting models best matched to the experimental question at hand.
Collapse
Affiliation(s)
- Joanna L Jankowsky
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Neurology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Neurosurgery, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Huffington Center on Aging, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Hui Zheng
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Huffington Center on Aging, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
37
|
Alaseem A, Alhazzani K, Dondapati P, Alobid S, Bishayee A, Rathinavelu A. Matrix Metalloproteinases: A challenging paradigm of cancer management. Semin Cancer Biol 2017; 56:100-115. [PMID: 29155240 DOI: 10.1016/j.semcancer.2017.11.008] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 11/15/2017] [Accepted: 11/15/2017] [Indexed: 12/11/2022]
Abstract
Matrix metalloproteinases (MMPs) are members of zinc-dependent endopeptidases implicated in a variety of physiological and pathological processes. Over the decades, MMPs have been studied for their role in cancer progression, migration, and metastasis. As a result, accumulated evidence of MMPs incriminating role has made them an attractive therapeutic target. Early generations of broad-spectrum MMP inhibitors exhibited potent inhibitory activities, which subsequently led to clinical trials. Unexpectedly, these trials failed to meet the desired goals, mainly due to the lack of efficacy, poor oral bioavailability, and toxicity. In this review, we discuss the regulatory role of MMPs in cancer progression, current strategies in targeting MMPs for cancer treatment including prodrug design and tumor imaging, and therapeutic value of MMPs as biomarkers in breast, lung, and prostate cancers.
Collapse
Affiliation(s)
- Ali Alaseem
- Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; College of Medicine, Al Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Khalid Alhazzani
- Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Priya Dondapati
- Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Saad Alobid
- Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | - Appu Rathinavelu
- Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
| |
Collapse
|
38
|
Rocca A, Tafuri D, Paccone M, Giuliani A, Zamboli AGI, Surfaro G, Paccone A, Compagna R, Amato M, Serra R, Amato B. Cell Based Therapeutic Approach in Vascular Surgery: Application and Review. Open Med (Wars) 2017; 12:308-322. [PMID: 29071303 PMCID: PMC5651406 DOI: 10.1515/med-2017-0045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 08/16/2017] [Indexed: 01/14/2023] Open
Abstract
Multipotent stem cells - such as mesenchymal stem/stromal cells and stem cells derived from different sources like vascular wall are intensely studied to try to rapidly translate their discovered features from bench to bedside. Vascular wall resident stem cells recruitment, differentiation, survival, proliferation, growth factor production, and signaling pathways transduced were analyzed. We studied biological properties of vascular resident stem cells and explored the relationship from several factors as Matrix Metalloproteinases (MMPs) and regulations of biological, translational and clinical features of these cells. In this review we described a translational and clinical approach to Adult Vascular Wall Resident Multipotent Vascular Stem Cells (VW-SCs) and reported their involvement in alternative clinical approach as cells based therapy in vascular disease like arterial aneurysms or peripheral arterial obstructive disease.
Collapse
Affiliation(s)
- Aldo Rocca
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, ItalyVia Sergio Pansini, 80131Naples, Italy
| | - Domenico Tafuri
- Department of Sport Sciences and Wellness, University of Naples “Parthenope”, Naples, Italy
| | - Marianna Paccone
- Department of Medicine and Health Sciences Vincenzo Tiberio, University of Molise, Campobasso, Italy
| | - Antonio Giuliani
- A.O.R.N. A. Cardarelli Hepatobiliary and Liver Transplatation Center, Naples, Italy
| | | | - Giuseppe Surfaro
- Antonio Cardarelli Hospital, General Surgery Unit, Campobasso, Italy
| | - Andrea Paccone
- Department of Medicine and Health Sciences Vincenzo Tiberio, University of Molise, Campobasso, Italy
| | - Rita Compagna
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Maurizo Amato
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Raffaele Serra
- Department of Medical and Surgical Sciences, University of Catanzaro, Catanzaro, Italy
| | - Bruno Amato
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
39
|
Cortes AL, Gonsalez SR, Rioja LS, Oliveira SSC, Santos ALS, Prieto MC, Melo PA, Lara LS. Protective outcomes of low-dose doxycycline on renal function of Wistar rats subjected to acute ischemia/reperfusion injury. Biochim Biophys Acta Mol Basis Dis 2017; 1864:102-114. [PMID: 28987762 DOI: 10.1016/j.bbadis.2017.10.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 08/08/2017] [Accepted: 10/03/2017] [Indexed: 02/06/2023]
Abstract
Renal ischemia-reperfusion injury (IRI) is a major cause of acute renal failure. Doxycycline (Dc) belongs to the tetracycline-class of antibiotics with demonstrated beneficial molecular effects in the brain and heart, mainly through matrix metalloproteinases inhibition (MMP). However, Dc protection of renal function has not been demonstrated. We determined whether low doses of Dc would prevent decreases in glomerular filtration rate (GFR) and maintain tubular Na+ handling in Wistar rats subjected to kidney I/R. Male Wistar rats underwent bilateral kidney ischemia for 30min followed by 24h reperfusion (I/R). Doxycycline (1, 3, and 10mg/kg, i.p.) was administered 2h before surgery. Untreated I/R rats showed a 250% increase in urine volume and proteinuria, a 60% reduction in GFR, accumulation of urea-nitrogen in the blood, and a 60% decrease in the fractional Na+ excretion due to unbalanced Na+ transporter activity. Treatment with Dc 3mg/kg maintained control levels of urine volume, proteinuria, GFR, blood urea-nitrogen, fractional Na+ excretion, and equilibrated Na+ transporter activities. The Dc protection effects on renal function were associated with kidney structure preservation and prevention of TGFβ and fibronectin deposition. In vitro, total MMP activity was augmented in I/R and inhibited by 25 and 50μM Dc. In vivo, I/R augmented MMP-2 and -9 protein content without changing their activities. Doxycycline treatment downregulated total MMP activity and MMP-2 and -9 protein content. Our results suggest that treatment with low dose Dc protects from IRI, thereby preserving kidney function.
Collapse
Affiliation(s)
- Aline L Cortes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sabrina R Gonsalez
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lilimar S Rioja
- Departamento de Patologia e Laboratórios, Universidade do Estado do Rio de Janeiro, Brazil
| | - Simone S C Oliveira
- Departamento de Microbiologia Geral, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - André L S Santos
- Departamento de Microbiologia Geral, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Minolfa C Prieto
- Department of Physiology, School of Medicine, Tulane University School of Medicine, New Orleans, LA, USA; Tulane Hypertension and Renal Center of Excellence, Tulane University, New Orleans, LA, USA
| | - Paulo A Melo
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucienne S Lara
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
40
|
Atchison L, Zhang H, Cao K, Truskey GA. A Tissue Engineered Blood Vessel Model of Hutchinson-Gilford Progeria Syndrome Using Human iPSC-derived Smooth Muscle Cells. Sci Rep 2017; 7:8168. [PMID: 28811655 PMCID: PMC5557922 DOI: 10.1038/s41598-017-08632-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/12/2017] [Indexed: 01/30/2023] Open
Abstract
Hutchison-Gilford Progeria Syndrome (HGPS) is a rare, accelerated aging disorder caused by nuclear accumulation of progerin, an altered form of the Lamin A gene. The primary cause of death is cardiovascular disease at about 14 years. Loss and dysfunction of smooth muscle cells (SMCs) in the vasculature may cause defects associated with HGPS. Due to limitations of 2D cell culture and mouse models, there is a need to develop improved models to discover novel therapeutics. To address this need, we produced a functional three-dimensional model of HGPS that replicates an arteriole-scale tissue engineered blood vessel (TEBV) using induced pluripotent stem cell (iPSC)-derived SMCs from an HGPS patient. To isolate the effect of the HGPS iSMCs, the endothelial layer consisted of human cord blood-derived endothelial progenitor cells (hCB-EPCs) from a separate, healthy donor. TEBVs fabricated from HGPS iSMCs and hCB-EPCs show reduced vasoactivity, increased medial wall thickness, increased calcification and apoptosis relative to TEBVs fabricated from normal iSMCs or primary MSCs. Additionally, treatment of HGPS TEBVs with the proposed therapeutic Everolimus, increases HGPS TEBV vasoactivity and increases iSMC differentiation in the TEBVs. These results show the ability of this iPSC-derived TEBV to reproduce key features of HGPS and respond to drugs.
Collapse
Affiliation(s)
- Leigh Atchison
- Department of Biomedical Engineering at Duke University, Durham, NC, 27708, United States
| | - Haoyue Zhang
- Department of Cell Biology and Molecular Genetics at University of Maryland, College Park, MD, 20742, United States
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics at University of Maryland, College Park, MD, 20742, United States
| | - George A Truskey
- Department of Biomedical Engineering at Duke University, Durham, NC, 27708, United States.
| |
Collapse
|
41
|
Yu M, Chen C, Cao Y, Qi R. Inhibitory effects of doxycycline on the onset and progression of abdominal aortic aneurysm and its related mechanisms. Eur J Pharmacol 2017; 811:101-109. [PMID: 28545777 DOI: 10.1016/j.ejphar.2017.05.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 03/22/2017] [Accepted: 05/22/2017] [Indexed: 11/28/2022]
Abstract
The objective of this study was to investigate whether doxycycline (DOX) given at different doses and via different administration routes had protective or therapeutic effects on abdominal aortic aneurysm (AAA) induced by elastase in mice. Moreover, the anti-AAA mechanism of DOX was studied in TNF-α-stimulated vascular smooth muscle cell (VSMC) in vitro. For in vivo study, either daily administration of 30mg/kg of DOX by gavage or intraperitoneal injection of 15mg/kg DOX every other day for 14 days significantly prevented the development of AAA at its early stage. Further study showed that intraperitoneal injection of 15mg/kg DOX every other day for 7 times in total could also cure the established AAA. In vitro study showed that treating VSMCs with TNF-α together with DOX remarkably inhibited the expressions and activities of MMPs (MMP-2 and MMP-9), significantly suppressed the activation of protein kinase B (AKT) signaling pathway and mitogen-activated protein kinases (MAPKs) signal proteins, including extracellular signal-regulated kinase (ERK), c-Jun amino-terminal kinases (JNK) and p38, and downregulated mRNA levels of interleukin-6 (IL-6) and monocyte chemotactic protein 1 (MCP-1), and significantly upregulated mRNA levels of transforming growth factor beta (TGF-β), heme oxygenase 1 (HO-1) and superoxide dismutase 1 (SOD-1), indicating that DOX inhibits activities of MMPs through reducing oxidative stress, suppressing MAPKs and AKT signaling pathways and ameliorating inflammation in VSMCs, and therefore, exerts preventive as well as therapeutic effects on AAA.
Collapse
Affiliation(s)
- Maomao Yu
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Peking University, Beijing 100191, China; Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing 100191, China
| | - Cong Chen
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Peking University, Beijing 100191, China; Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing 100191, China
| | - Yini Cao
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Peking University, Beijing 100191, China; Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing 100191, China
| | - Rong Qi
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Peking University, Beijing 100191, China; Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing 100191, China.
| |
Collapse
|
42
|
Sivaraman B, Swaminathan G, Moore L, Fox J, Seshadri D, Dahal S, Stoilov I, Zborowski M, Mecham R, Ramamurthi A. Magnetically-responsive, multifunctional drug delivery nanoparticles for elastic matrix regenerative repair. Acta Biomater 2017; 52:171-186. [PMID: 27884774 DOI: 10.1016/j.actbio.2016.11.048] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/14/2016] [Accepted: 11/20/2016] [Indexed: 12/23/2022]
Abstract
Arresting or regressing growth of abdominal aortic aneurysms (AAAs), localized expansions of the abdominal aorta are contingent on inhibiting chronically overexpressed matrix metalloproteases (MMPs)-2 and -9 that disrupt elastic matrix within the aortic wall, concurrent with providing a stimulus to augmenting inherently poor auto-regeneration of these matrix structures. In a recent study we demonstrated that localized, controlled and sustained delivery of doxycycline (DOX; a tetracycline-based antibiotic) from poly(lactic-co-glycolic acid) nanoparticles (PLGA NPs), enhances elastic matrix deposition and MMP-inhibition at a fraction of the therapeutically effective oral dose. The surface functionalization of these NPs with cationic amphiphiles, which enhances their arterial uptake, was also shown to have pro-matrix regenerative and anti-MMP effects independent of the DOX. Based on the hypothesis that the incorporation of superparamagnetic iron oxide NPs (SPIONs) within these PLGA NPs would enhance their targetability to the AAA site under an applied external magnetic field, we sought to evaluate the functional effects of NPs co-encapsulating DOX and SPIONs (DOX-SPION NPs) on elastic matrix regeneration and MMP synthesis/activity in vitro within aneurysmal smooth muscle cell (EaRASMC) cultures. The DOX-SPION NPs were mobile under an applied external magnetic field, while enhancing elastic matrix deposition 1.5-2-fold and significantly inhibiting MMP-2 synthesis and MMP-2 and -9 activities, compared to NP-untreated control cultures. These results illustrate that the multifunctional benefits of NPs are maintained following SPION co-incorporation. Additionally, preliminary studies carried out demonstrated enhanced targetability of SPION-loaded NPs within proteolytically-disrupted porcine carotid arteries ex vivo, under the influence of an applied external magnetic field. Thus, this dual-agent loaded NP system proffers a potential non-surgical option for treating small growing AAAs, via controlled and sustained drug release from multifunctional, targetable nanocarriers. STATEMENT OF SIGNIFICANCE Proactive screening of high risk elderly patients now enables early detection of abdominal aortic aneurysms (AAAs). There are no established drug-based therapeutic alternatives to surgery for AAAs, which is unsuitable for many elderly patients, and none which can achieve restore disrupted and lost elastic matrix in the AAA wall, which is essential to achieve growth arrest or regression. We have developed a first generation design of polymer nanoparticles (NPs) for AAA tissue localized delivery of doxycycline, a modified tetracycline drug at low micromolar doses at which it provides both pro-elastogenic and anti-proteolytic benefits that can augment elastic matrix regenerative repair. The nanocarriers themselves are also uniquely chemically functionalized on their surface to also provide them pro-elastin-regenerative & anti-matrix degradative properties. To provide an active driving force for efficient uptake of intra-lumenally infused NPs to the AAA wall, in this work, we have rendered our polymer NPs mobile in an applied magnetic field via co-incorporation of super-paramagnetic iron oxide NPs. We demonstrate that such modifications significantly improve wall uptake of the NPs with no significant changes to their physical properties and regenerative benefits. Such NPs can potentially stimulate structural repair in the AAA wall following one time infusion to delay or prevent AAA growth to rupture. The therapy can provide a non-surgical treatment option for high risk AAA patients.
Collapse
|
43
|
Jennewine B, Fox J, Ramamurthi A. Cathepsin K-targeted sub-micron particles for regenerative repair of vascular elastic matrix. Acta Biomater 2017; 52:60-73. [PMID: 28087488 PMCID: PMC6361138 DOI: 10.1016/j.actbio.2017.01.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 11/14/2016] [Accepted: 01/10/2017] [Indexed: 12/20/2022]
Abstract
Abdominal Aortic Aneurysms (AAA) involve slow dilation and weakening of the aortic wall due to breakdown of structural matrix components, such as elastic fibers by chronically overexpressed matrix metalloproteinases (MMPs), primarily, MMPs-2 and -9. Auto-regenerative repair of disrupted elastic fibers by smooth muscle cells (SMCs) at the AAA site is intrinsically poor and together with chronic proteolysis prevents restoration of elastin homeostasis, necessary to enable AAA growth arrest or regression to a healthy state. Oral doxycycline (DOX) therapy can inhibit MMPs to slow AAA growth, but has systemwide side-effects and inhibits new elastin deposition within AAA tissue, diminishing prospects for restoring elastin homeostasis preventing the arrest/regression of AAA growth. We have thus developed cationic amphiphile (DMAB)-modified submicron particles (SMPs) that uniquely exhibit pro-elastogenic and anti-proteolytic properties, separate from similar effects of the encapsulated drug. These SMPs can enable sustained, low dose DOX delivery within AAA tissue to augment elastin regenerative repair. To provide greater specificity of SMP targeting, we have conjugated the DOX-SMP surface with an antibody against cathepsin K, a lysosomal protease that is highly overexpressed within AAA tissue. We have determined conditions for efficient cathepsin K Ab conjugation onto the SMPs, improved SMP binding to aneurysmal SMCs in culture and to injured vessel walls ex vivo, conjugation did not affect DOX release from the SMPs, and improved pro-elastogenic and anti-proteolytic effects due to the SMPs likely due to their increased proximity to cells via binding. Our study results suggest that cathepsin K Ab conjugation is a useful targeting modality for our pro-regenerative SMPs. Future studies will investigate SMP retention and biodistribution following targeting to induced AAAs in rat models through intravenous or catheter-based aortal infusion and thereafter their efficacy for regenerative elastic matrix repair in the AAA wall. STATEMENT OF SIGNIFICANCE Proactive screening of high risk elderly patients now enables early detection of Abdominal Aortic Aneurysms (AAAs). Current management of small, growing AAAs is limited to passive, imaging based growth monitoring. There are also no established drug-based therapeutic alternatives to surgery for AAAs, which is unsuitable for many elderly patients, and none which can achieve restore disrupted and lost elastic matrix in the AAA wall, which is essential to achieve growth arrest or regression. We seek to test the feasibility of a regenerative therapy based on localized, one time delivery of drug-releasing Sub-Micron-sized drug delivery polymer Particles (SMPs) that are also uniquely chemically functionalized on their surface to also provide them pro-elastin-regenerative & anti-matrix degradative properties, and also conjugated with antibodies targeting cathepsin K, an elastolytic enzyme that is highly overexpressed in AAA tissues; the latter serves as a modality to enable targeted binding of the SMPs to the AAA wall following intravenous infusion, or intraoartal, catheter-based delivery. Such SMPs can potentially stimulate structural repair in the AAA wall following one time infusion to delay or prevent AAA growth to rupture. The therapy can provide a non-surgical treatment option for high risk AAA patients.
Collapse
Affiliation(s)
- Brenton Jennewine
- Department of Biomedical Engineering, The Cleveland Clinic, 9500 Euclid Avenue, ND 20, Cleveland, OH 44195, USA; Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Jonathan Fox
- Department of Biomedical Engineering, The Cleveland Clinic, 9500 Euclid Avenue, ND 20, Cleveland, OH 44195, USA; Department of Chemical and Biomedical Engineering, Cleveland State University, 2121 Euclid Avenue, Cleveland, OH 44115, USA
| | - Anand Ramamurthi
- Department of Biomedical Engineering, The Cleveland Clinic, 9500 Euclid Avenue, ND 20, Cleveland, OH 44195, USA; Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| |
Collapse
|
44
|
Costanza B, Umelo IA, Bellier J, Castronovo V, Turtoi A. Stromal Modulators of TGF-β in Cancer. J Clin Med 2017; 6:jcm6010007. [PMID: 28067804 PMCID: PMC5294960 DOI: 10.3390/jcm6010007] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/19/2016] [Accepted: 12/23/2016] [Indexed: 02/07/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is an intriguing cytokine exhibiting dual activities in malignant disease. It is an important mediator of cancer invasion, metastasis and angiogenesis, on the one hand, while it exhibits anti-tumor functions on the other hand. Elucidating the precise role of TGF-β in malignant development and progression requires a better understanding of the molecular mechanisms involved in its tumor suppressor to tumor promoter switch. One important aspect of TGF-β function is its interaction with proteins within the tumor microenvironment. Several stromal proteins have the natural ability to interact and modulate TGF-β function. Understanding the complex interplay between the TGF-β signaling network and these stromal proteins may provide greater insight into the development of novel therapeutic strategies that target the TGF-β axis. The present review highlights our present understanding of how stroma modulates TGF-β activity in human cancers.
Collapse
Affiliation(s)
- Brunella Costanza
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, 4000 Liege, Belgium.
| | - Ijeoma Adaku Umelo
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, 4000 Liege, Belgium.
| | - Justine Bellier
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, 4000 Liege, Belgium.
| | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, 4000 Liege, Belgium.
| | - Andrei Turtoi
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, 4000 Liege, Belgium.
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université Montpellier, Institut Régional du Cancer de Montpellier, 34298 Montpellier, France.
| |
Collapse
|
45
|
Jung JJ, Razavian M, Kim HY, Ye Y, Golestani R, Toczek J, Zhang J, Sadeghi MM. Matrix metalloproteinase inhibitor, doxycycline and progression of calcific aortic valve disease in hyperlipidemic mice. Sci Rep 2016; 6:32659. [PMID: 27619752 PMCID: PMC5020643 DOI: 10.1038/srep32659] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/12/2016] [Indexed: 12/18/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is the most common cause of aortic stenosis. Currently, there is no non-invasive medical therapy for CAVD. Matrix metalloproteinases (MMPs) are upregulated in CAVD and play a role in its pathogenesis. Here, we evaluated the effect of doxycycline, a nonselective MMP inhibitor on CAVD progression in the mouse. Apolipoprotein (apo)E−/− mice (n = 20) were fed a Western diet (WD) to induce CAVD. After 3 months, half of the animals was treated with doxycycline, while the others continued WD alone. After 6 months, we evaluated the effect of doxycycline on CAVD progression by echocardiography, MMP-targeted micro single photon emission computed tomography (SPECT)/computed tomography (CT), and tissue analysis. Despite therapeutic blood levels, doxycycline had no significant effect on MMP activation, aortic valve leaflet separation or flow velocity. This lack of effect on in vivo images was confirmed on tissue analysis which showed a similar level of aortic valve gelatinase activity, and inflammation between the two groups of animals. In conclusion, doxycycline (100 mg/kg/day) had no effect on CAVD progression in apoE−/− mice with early disease. Studies with more potent and specific inhibitors are needed to establish any potential role of MMP inhibition in CAVD development and progression.
Collapse
Affiliation(s)
- Jae-Joon Jung
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, United States.,VA Connecticut Healthcare System, West Haven, CT, United States
| | - Mahmoud Razavian
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, United States.,VA Connecticut Healthcare System, West Haven, CT, United States
| | - Hye-Yeong Kim
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, United States.,VA Connecticut Healthcare System, West Haven, CT, United States
| | - Yunpeng Ye
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, United States.,VA Connecticut Healthcare System, West Haven, CT, United States
| | - Reza Golestani
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, United States.,VA Connecticut Healthcare System, West Haven, CT, United States
| | - Jakub Toczek
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, United States.,VA Connecticut Healthcare System, West Haven, CT, United States
| | - Jiasheng Zhang
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, United States.,VA Connecticut Healthcare System, West Haven, CT, United States
| | - Mehran M Sadeghi
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, United States.,VA Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
46
|
Parasaram V, Nosoudi N, LeClair RJ, Binks A, Vyavahare N. Targeted drug delivery to emphysematous lungs: Inhibition of MMPs by doxycycline loaded nanoparticles. Pulm Pharmacol Ther 2016; 39:64-73. [PMID: 27354173 DOI: 10.1016/j.pupt.2016.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/16/2016] [Accepted: 06/19/2016] [Indexed: 01/03/2023]
Affiliation(s)
| | - Nasim Nosoudi
- Department of Bioengineering, Clemson University, SC, USA
| | - Renee J LeClair
- University of South Carolina School of Medicine, Greenville, SC, USA
| | - Andrew Binks
- University of South Carolina School of Medicine, Greenville, SC, USA
| | | |
Collapse
|
47
|
Jacob T, Hingorani A, Ascher E. Role of Apoptosis and Proteolysis in the Pathogenesis of Iliac Artery Aneurysms. Vascular 2016; 13:34-42. [PMID: 15895673 DOI: 10.1258/rsmvasc.13.1.34] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The objective of this study was to investigate the role of inflammation, programmed cell death, its molecular modulators, and proteolysis in the pathogenesis of iliac artery aneurysms (IAAs). Nineteen IAA specimens were obtained from patients undergoing elective surgical repair. All were males with ages ranging from 55 to 85 years (mean 73 years). Controls were iliac arteries ( n = 6) retrieved from surgical patients without aneurysmal disease. Standard histochemical techniques were used to assess elastic lamellae fragmentation and inflammatory infiltrate in aneurysmal and normal tissues. Identification of different types of cells in the aneurysm wall and detection of death-promoting molecules, Fas, p53, perforin, apoptosis-mediating bcl-2 family proteins, apoptotic death substrate, and poly(adenosine diphosphate–ribose) polymerase were performed immunohistochemically. Apoptosis was detected by terminal deoxynucleotidyl transferase–mediated digoxigenin–deoxyuridine triphosphate nick end-labeling (TUNEL) assay and caspase activity. Proteolytic activity was determined by 10% gelatin gel zymography. There is a conspicuous disruption and fragmentation of elastic lamellae in IAAs compared with normal arteries. Increased gelatinolytic activity was observed at 92, 72, and 67 kDa in the aneurysmal tissues. There was a significant loss of vascular smooth muscle cells (VSMCs) in the IAA walls compared with normal arteries ( p < .02). Large numbers of inflammatory cells were observed in the IAA specimens ( p = .01). Only aneurysmal arteries showed CD8+ T cells expressing death-promoting molecules. CD3+, CD8+, CD20+, CD30+, and CD68+ immunoreactive cells were significantly more prominent in the aneurysmal tissues than in the control arteries. There was a significant increase in the number of cells undergoing apoptosis in aneurysmal tissue than in the normal vessels ( p < .02), as well as in the expression of bax, p53, CPP-32, and Fas. Apoptotic cells and proapoptotic molecules predominantly localized to the inflammatory infiltrate. VSMC apoptosis was significant in IAAs. The data confirm the architectural disruption of the IAA wall and illustrate an apparent biologic response involving inflammatory infiltrate, apoptosis, and signaling molecules capable of initiating cell death. In addition to compromising the mechanical integrity of the vessel wall, VSMC loss may contribute to imbalance in the protein profile, accelerating extracellular matrix degradation that could favor IAA development.
Collapse
Affiliation(s)
- Theresa Jacob
- Division of Vascular Surgery, Department of Surgery, Maimonides Medical Center, Brooklyn, NY, USA
| | | | | |
Collapse
|
48
|
Rentschler ME, Baxter B. Medical Therapy Approach for Treating Abdominal Aortic Aneurysm. Vascular 2016; 15:361-5. [DOI: 10.2310/6670.2007.00055] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Abdominal aortic aneurysm (AAA) is a common and deadly problem. The aortic diameter increases in association with a complex remodeling process that includes changes in the structure and content of key proteins, elastin and collagen. As these changes occur, the tissue mechanical properties also change. The natural history of AAA is progressive enlargement to a point of mechanical tissue failure, typically followed by death. Currently, the marker used to predict the risk of impending rupture is the largest transverse diameter. After reaching a diameter threshold of 5.5 cm, the aneurysm is surgically repaired. This criterion does not consider any patient-specific information or the known heterogeneity of the aneurysm that may, in some cases, lead to rupture before the aneurysm reaches the standard intervention threshold. Conversely, in many patients, continued observation beyond this threshold is safe. Although no medical treatment is yet approved, doxycycline has been shown to greatly reduce aortic aneurysm growth in animal models and has been shown to slow growth in several small clinical trials. Although larger prospective randomized trials are needed, one unknown is what effect doxycycline has on the structural integrity of the aortic wall. That is, does slowed aneurysm growth by doxycycline treatments, in fact, prevent rupture, or does the wall continue to weaken and the aneurysm instead ruptures at a smaller diameter? Research has begun to answer these questions before a large clinical trial begins.
Collapse
Affiliation(s)
- Mark E. Rentschler
- *Department of Surgery, University of Nebraska Medical Center, Omaha, NE
| | - B.Timothy Baxter
- *Department of Surgery, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
49
|
Ordonez AA, Tasneen R, Pokkali S, Xu Z, Converse PJ, Klunk MH, Mollura DJ, Nuermberger EL, Jain SK. Mouse model of pulmonary cavitary tuberculosis and expression of matrix metalloproteinase-9. Dis Model Mech 2016; 9:779-88. [PMID: 27482816 PMCID: PMC4958312 DOI: 10.1242/dmm.025643] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/20/2016] [Indexed: 01/12/2023] Open
Abstract
Cavitation is a key pathological feature of human tuberculosis (TB), and is a well-recognized risk factor for transmission of infection, relapse after treatment and the emergence of drug resistance. Despite intense interest in the mechanisms underlying cavitation and its negative impact on treatment outcomes, there has been limited study of this phenomenon, owing in large part to the limitations of existing animal models. Although cavitation does not occur in conventional mouse strains after infection with Mycobacterium tuberculosis, cavitary lung lesions have occasionally been observed in C3HeB/FeJ mice. However, to date, there has been no demonstration that cavitation can be produced consistently enough to support C3HeB/FeJ mice as a new and useful model of cavitary TB. We utilized serial computed tomography (CT) imaging to detect pulmonary cavitation in C3HeB/FeJ mice after aerosol infection with M. tuberculosis Post-mortem analyses were performed to characterize lung lesions and to localize matrix metalloproteinases (MMPs) previously implicated in cavitary TB in situ A total of 47-61% of infected mice developed cavities during primary disease or relapse after non-curative treatments. Key pathological features of human TB, including simultaneous presence of multiple pathologies, were noted in lung tissues. Optical imaging demonstrated increased MMP activity in TB lesions and MMP-9 was significantly expressed in cavitary lesions. Tissue MMP-9 activity could be abrogated by specific inhibitors. In situ, three-dimensional analyses of cavitary lesions demonstrated that 22.06% of CD11b+ signal colocalized with MMP-9. C3HeB/FeJ mice represent a reliable, economical and tractable model of cavitary TB, with key similarities to human TB. This model should provide an excellent tool to better understand the pathogenesis of cavitation and its effects on TB treatments.
Collapse
Affiliation(s)
- Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Rokeya Tasneen
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Supriya Pokkali
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ziyue Xu
- Center for Infectious Disease Imaging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Paul J Converse
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mariah H Klunk
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Daniel J Mollura
- Center for Infectious Disease Imaging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eric L Nuermberger
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
50
|
Wassenaar JW, Braden RL, Osborn KG, Christman KL. Modulating In Vivo Degradation Rate of Injectable Extracellular Matrix Hydrogels. J Mater Chem B 2016; 4:2794-2802. [PMID: 27563436 PMCID: PMC4993464 DOI: 10.1039/c5tb02564h] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Extracellular matrix (ECM) derived hydrogels are increasingly used as scaffolds to stimulate endogenous repair. However, few studies have examined how altering the degradation rates of these materials affect cellular interaction in vivo. This study sought to examine how crosslinking or matrix metalloproteinase (MMP) inhibition by doxycycline could be employed to modulate the degradation rate of an injectable hydrogel derived from decellularized porcine ventricular myocardium. While both approaches were effective in reducing degradation in vitro, only doxycycline significantly prolonged hydrogel degradation in vivo without affecting material biocompatibility. In addition, unlike crosslinking, incorporation of doxycycline into the hydrogel did not affect mechanical properties. Lastly, the results of this study highlighted the need for development of novel crosslinkers for in situ modification of injectable ECM-derived hydrogels, as none of the crosslinking agents investigated in this study were both biocompatible and effective.
Collapse
Affiliation(s)
- Jean W. Wassenaar
- Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California, San Diego
| | - Rebecca L. Braden
- Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California, San Diego
| | - Kent G. Osborn
- Office of Animal Research, University of California, San Diego
| | - Karen L. Christman
- Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California, San Diego
| |
Collapse
|