1
|
Kang JH, Uddin N, Kim S, Zhao Y, Yoo KC, Kim MJ, Hong SA, Bae S, Lee JY, Shin I, Jin YW, O'Hagan HM, Yi JM, Lee SJ. Tumor-intrinsic role of ICAM-1 in driving metastatic progression of triple-negative breast cancer through direct interaction with EGFR. Mol Cancer 2024; 23:230. [PMID: 39415210 PMCID: PMC11481280 DOI: 10.1186/s12943-024-02150-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024] Open
Abstract
Triple-negative breast cancer (TNBC), the most aggressive subtype, presents a critical challenge due to the absence of approved targeted therapies. Hence, there is an urgent need to identify effective therapeutic targets for this condition. While epidermal growth factor receptor (EGFR) is prominently expressed in TNBC and recognized as a therapeutic target, anti-EGFR therapies have yet to gain approval for breast cancer treatment due to their associated side effects and limited efficacy. Here, we discovered that intercellular adhesion molecule-1 (ICAM-1) exhibits elevated expression levels in metastatic breast cancer and serves as a pivotal binding adaptor for EGFR activation, playing a crucial role in malignant progression. The activation of EGFR by tumor-expressed ICAM-1 initiates biased signaling within the JAK1/STAT3 pathway, consequently driving epithelial-to-mesenchymal transition and facilitating heightened metastasis without influencing tumor growth. Remarkably, ICAM-1-neutralizing antibody treatment significantly suppressed cancer metastasis in a breast cancer orthotopic xenograft mouse model. In conclusion, our identification of ICAM-1 as a novel tumor intrinsic regulator of EGFR activation offers valuable insights for the development of TNBC-specific anti-EGFR therapies.
Collapse
Affiliation(s)
- Jae-Hyeok Kang
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, South Korea
| | - Nizam Uddin
- Center for Cell Analysis & Modeling, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Seungmo Kim
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, South Korea
| | - Yi Zhao
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, South Korea
| | - Ki-Chun Yoo
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, South Korea
| | - Min-Jung Kim
- Fibrosis and Cancer Targeting Biotechnology (FNCT BIOTECH), Toegye-Ro 36 Gil, Seoul, 04626, South Korea
| | - Sung-Ah Hong
- Genomic Medicine Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Sangsu Bae
- Department of Biochemistry and Molecular Biology, College of Medicine, Seoul National University, Seoul, 03080, South Korea
| | - Jeong-Yeon Lee
- Department of Pathology, College of Medicine, Hanyang University, Seoul, 04763, South Korea
| | - Incheol Shin
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, South Korea
| | - Young Woo Jin
- Fibrosis and Cancer Targeting Biotechnology (FNCT BIOTECH), Toegye-Ro 36 Gil, Seoul, 04626, South Korea
| | - Heather M O'Hagan
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Joo Mi Yi
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Bloomington, IN, 47405, USA.
- Department of Microbiology and Immunology, College of Medicine, Inje University, Busan, 47392, South Korea.
| | - Su-Jae Lee
- Fibrosis and Cancer Targeting Biotechnology (FNCT BIOTECH), Toegye-Ro 36 Gil, Seoul, 04626, South Korea.
| |
Collapse
|
2
|
Ramírez-Carracedo R, Hernández I, Moreno-Gómez-Toledano R, Díez-Mata J, Tesoro L, González-Cucharero C, Jiménez-Guirado B, Alcharani N, Botana L, Saura M, Zamorano JL, Zaragoza C. NOS3 prevents MMP-9, and MMP-13 induced extracellular matrix proteolytic degradation through specific microRNA-targeted expression of extracellular matrix metalloproteinase inducer in hypertension-related atherosclerosis. J Hypertens 2024; 42:685-693. [PMID: 38406874 PMCID: PMC10906209 DOI: 10.1097/hjh.0000000000003679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 01/08/2024] [Accepted: 01/22/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND Endothelial nitric oxide synthase (NOS3) elicits atheroprotection by preventing extracellular matrix (ECM) proteolytic degradation through inhibition of extracellular matrix metalloproteinase inducer (EMMPRIN) and collagenase MMP-13 by still unknown mechanisms. METHODS C57BL/6 mice lacking ApoE , NOS3, and/or MMP13 were fed with a high-fat diet for 6 weeks. Entire aortas were extracted and frozen to analyze protein and nucleic acid expression. Atherosclerotic plaques were detected by ultrasound imaging, Oil Red O (ORO) staining, and Western Blot. RNA-seq and RT-qPCR were performed to evaluate EMMPRIN, MMP-9, and EMMPRIN-targeting miRNAs. Mouse aortic endothelial cells (MAEC) were incubated to assess the role of active MMP-13 over MMP-9. One-way ANOVA or Kruskal-Wallis tests were performed to determine statistical differences. RESULTS Lack of NOS3 in ApoE null mice fed with a high-fat diet increased severe plaque accumulation, vessel wall widening, and high mortality, along with EMMPRIN-induced expression by upregulation of miRNAs 46a-5p and 486-5p. However, knocking out MMP-13 in ApoE/NOS3 -deficient mice was sufficient to prevent mortality (66.6 vs. 26.6%), plaque progression (23.1 vs. 8.8%), and MMP-9 expression, as confirmed in murine aortic endothelial cell (MAEC) cultures, in which MMP-9 was upregulated by incubation with active recombinant MMP-13, suggesting MMP-9 as a new target of MMP-13 in atherosclerosis. CONCLUSION We describe a novel mechanism by which the absence of NOS3 may worsen atherosclerosis through EMMPRIN-induced ECM proteolytic degradation by targeting the expression of miRNAs 146a-5p and 485-5p. Focusing on NOS3 regulation of ECM degradation could be a promising approach in the management of atherosclerosis.
Collapse
Affiliation(s)
- Rafael Ramírez-Carracedo
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
| | - Ignacio Hernández
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos
| | - Rafael Moreno-Gómez-Toledano
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
- Universidad de Alcalá, Unidad de Fisiología, Departamento de Biología de Sistemas, Alcalá de Henares
| | - Javier Díez-Mata
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
| | - Laura Tesoro
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
| | - Claudia González-Cucharero
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
| | - Beatriz Jiménez-Guirado
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
| | - Nunzio Alcharani
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
| | - Laura Botana
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
| | - Marta Saura
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos
- Universidad de Alcalá, Unidad de Fisiología, Departamento de Biología de Sistemas, Alcalá de Henares
| | - Jose L. Zamorano
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos
- Departamento de Cardiología, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, Spain
| | - Carlos Zaragoza
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos
| |
Collapse
|
3
|
Reventun P, Sánchez-Esteban S, Cook-Calvete A, Delgado-Marín M, Roza C, Jorquera-Ortega S, Hernandez I, Tesoro L, Botana L, Zamorano JL, Zaragoza C, Saura M. Endothelial ILK induces cardioprotection by preventing coronary microvascular dysfunction and endothelial-to-mesenchymal transition. Basic Res Cardiol 2023; 118:28. [PMID: 37452166 PMCID: PMC10348984 DOI: 10.1007/s00395-023-00997-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/13/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
Endothelial dysfunction is an early event in coronary microvascular disease. Integrin-linked kinase (ILK) prevents endothelial nitric oxide synthase (eNOS) uncoupling and, thus, endothelial dysfunction. However, the specific role of endothelial ILK in cardiac function remains to be fully elucidated. We hypothesised that endothelial ILK plays a crucial role in maintaining coronary microvascular function and contractile performance in the heart. We generated an endothelial cell-specific ILK conditional knock-out mouse (ecILK cKO) and investigated cardiovascular function. Coronary endothelial ILK deletion significantly impaired cardiac function: ejection fraction, fractional shortening and cardiac output decreased, whilst left ventricle diastolic internal diameter decreased and E/A and E/E' ratios increased, indicating not only systolic but also diastolic dysfunction. The functional data correlated with extensive extracellular matrix remodelling and perivascular fibrosis, indicative of adverse cardiac remodelling. Mice with endothelial ILK deletion suffered early ischaemic-like events with ST elevation and transient increases in cardiac troponins, which correlated with fibrotic remodelling. In addition, ecILK cKO mice exhibited many features of coronary microvascular disease: reduced cardiac perfusion, impaired coronary flow reserve and arterial remodelling with patent epicardial coronary arteries. Moreover, endothelial ILK deletion induced a moderate increase in blood pressure, but the antihypertensive drug Losartan did not affect microvascular remodelling whilst only partially ameliorated fibrotic remodelling. The plasma miRNA profile reveals endothelial-to-mesenchymal transition (endMT) as an upregulated pathway in endothelial ILK conditional KO mice. Our results show that endothelial cells in the microvasculature in endothelial ILK conditional KO mice underwent endMT. Moreover, endothelial cells isolated from these mice and ILK-silenced human microvascular endothelial cells underwent endMT, indicating that decreased endothelial ILK contributes directly to this endothelial phenotype shift. Our results identify ILK as a crucial regulator of microvascular endothelial homeostasis. Endothelial ILK prevents microvascular dysfunction and cardiac remodelling, contributing to the maintenance of the endothelial cell phenotype.
Collapse
Affiliation(s)
- P Reventun
- Facultad Medicina, Depto. Biología Sistemas (UD Fisiología), Universidad de Alcalá, IRYCIS, Mod 2 Planta 0, Ctra Madrid, Barcelona Km 33,500, Alcalá de Henares, Madrid, Spain
- School of Medicine, Department of Medicine, Cardiology Division, Johns Hopkins University, Baltimore, MD, United States
| | - S Sánchez-Esteban
- Facultad Medicina, Depto. Biología Sistemas (UD Fisiología), Universidad de Alcalá, IRYCIS, Mod 2 Planta 0, Ctra Madrid, Barcelona Km 33,500, Alcalá de Henares, Madrid, Spain
| | - A Cook-Calvete
- Facultad Medicina, Depto. Biología Sistemas (UD Fisiología), Universidad de Alcalá, IRYCIS, Mod 2 Planta 0, Ctra Madrid, Barcelona Km 33,500, Alcalá de Henares, Madrid, Spain
| | - M Delgado-Marín
- Facultad Medicina, Depto. Biología Sistemas (UD Fisiología), Universidad de Alcalá, IRYCIS, Mod 2 Planta 0, Ctra Madrid, Barcelona Km 33,500, Alcalá de Henares, Madrid, Spain
| | - C Roza
- Facultad Medicina, Depto. Biología Sistemas (UD Fisiología), Universidad de Alcalá, IRYCIS, Mod 2 Planta 0, Ctra Madrid, Barcelona Km 33,500, Alcalá de Henares, Madrid, Spain
| | - S Jorquera-Ortega
- Facultad Medicina, Depto. Biología Sistemas (UD Fisiología), Universidad de Alcalá, IRYCIS, Mod 2 Planta 0, Ctra Madrid, Barcelona Km 33,500, Alcalá de Henares, Madrid, Spain
| | - I Hernandez
- Unidad Mixta de Investigación Cardiovascular, Universidad Francisco de Vitoria, IRYCIS, Pozuelo de Alarcón, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - L Tesoro
- Unidad Mixta de Investigación Cardiovascular, Universidad Francisco de Vitoria, IRYCIS, Pozuelo de Alarcón, Madrid, Spain
| | - L Botana
- Unidad Mixta de Investigación Cardiovascular, Universidad Francisco de Vitoria, IRYCIS, Pozuelo de Alarcón, Madrid, Spain
| | - J L Zamorano
- Servicio Cardiología, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - C Zaragoza
- Unidad Mixta de Investigación Cardiovascular, Universidad Francisco de Vitoria, IRYCIS, Pozuelo de Alarcón, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - M Saura
- Facultad Medicina, Depto. Biología Sistemas (UD Fisiología), Universidad de Alcalá, IRYCIS, Mod 2 Planta 0, Ctra Madrid, Barcelona Km 33,500, Alcalá de Henares, Madrid, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| |
Collapse
|
4
|
Yoo K, Kang J, Choi M, Suh Y, Zhao Y, Kim M, Chang JH, Shim J, Yoon S, Kang S, Lee S. Soluble ICAM-1 a Pivotal Communicator between Tumors and Macrophages, Promotes Mesenchymal Shift of Glioblastoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2102768. [PMID: 34813169 PMCID: PMC8805565 DOI: 10.1002/advs.202102768] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/18/2021] [Indexed: 06/13/2023]
Abstract
Despite aggressive clinical treatment, recurrence of glioblastoma multiforme (GBM) is unavoidable, and the clinical outcome is still poor. A convincing explanation is the phenotypic transition of GBM cells upon aggressive treatment such as radiotherapy. However, the microenvironmental factors contributing to GBM recurrence after treatment remain unexplored. Here, it is shown that radiation-treated GBM cells produce soluble intercellular adhesion molecule-1 (sICAM-1) which stimulates the infiltration of macrophages, consequently enriching the tumor microenvironment with inflammatory macrophages. Acting as a paracrine factor, tumor-derived sICAM-1 induces macrophages to secrete wingless-type MMTV integration site family, member 3A (WNT3A), which promotes a mesenchymal shift of GBM cells. In addition, blockade of either sICAM-1 or WNT3A diminishes the harmful effect of radiation on tumor progression. Collectively, the findings indicate that cellular crosstalk between GBM and macrophage through sICAM-1-WNT3A oncogenic route is involved in the mesenchymal shift of GBM cells after radiation, and suggest that radiotherapy combined with sICAM-1 targeted inhibition would improve the clinical outcome of GBM patients.
Collapse
Affiliation(s)
- Ki‐Chun Yoo
- Department of Life ScienceResearch Institute for Natural SciencesHanyang UniversitySeoul04763Korea
- Department of Lymphoma and MyelomaDivision of Cancer MedicineCenter for Cancer Immunology ResearchThe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Jae‐Hyeok Kang
- Department of Life ScienceResearch Institute for Natural SciencesHanyang UniversitySeoul04763Korea
| | - Mi‐Young Choi
- Department of Life ScienceResearch Institute for Natural SciencesHanyang UniversitySeoul04763Korea
| | - Yongjoon Suh
- Department of Life ScienceResearch Institute for Natural SciencesHanyang UniversitySeoul04763Korea
| | - Yi Zhao
- Department of Life ScienceResearch Institute for Natural SciencesHanyang UniversitySeoul04763Korea
| | - Min‐Jung Kim
- Laboratory of Radiation Exposure & TherapeuticsNational Radiation Emergency Medical CenterKorea Institute of Radiological and Medical SciencesSeoul01812Korea
| | - Jong Hee Chang
- Department of NeurosurgeryBrain Tumor CenterSeverance HospitalYonsei University College of MedicineSeoul03722Korea
| | - Jin‐Kyoung Shim
- Department of NeurosurgeryBrain Tumor CenterSeverance HospitalYonsei University College of MedicineSeoul03722Korea
| | - Seon‐Jin Yoon
- Department of NeurosurgeryBrain Tumor CenterSeverance HospitalYonsei University College of MedicineSeoul03722Korea
| | - Seok‐Gu Kang
- Department of NeurosurgeryBrain Tumor CenterSeverance HospitalYonsei University College of MedicineSeoul03722Korea
| | - Su‐Jae Lee
- Department of Life ScienceResearch Institute for Natural SciencesHanyang UniversitySeoul04763Korea
| |
Collapse
|
5
|
Raffetto JD, Khalil RA. Mechanisms of Lower Extremity Vein Dysfunction in Chronic Venous Disease and Implications in Management of Varicose Veins. VESSEL PLUS 2021; 5. [PMID: 34250453 DOI: 10.20517/2574-1209.2021.16] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Chronic venous disease (CVD) is a common venous disorder of the lower extremities. CVD can be manifested as varicose veins (VVs), with dilated and tortuous veins, dysfunctional valves and venous reflux. If not adequately treated, VVs could progress to chronic venous insufficiency (CVI) and lead to venous leg ulcer (VLU). Predisposing familial and genetic factors have been implicated in CVD. Additional environmental, behavioral and dietary factors including sedentary lifestyle and obesity may also contribute to CVD. Alterations in the mRNA expression, protein levels and proteolytic activity of matrix metalloproteinases (MMPs) have been detected in VVs and VLU. MMP expression/activity can be modulated by venous hydrostatic pressure, hypoxia, tissue metabolites, and inflammation. MMPs in turn increase proteolysis of different protein substrates in the extracellular matrix particularly collagen and elastin, leading to weakening of the vein wall. MMPs could also promote venous dilation by increasing the release of endothelium-derived vasodilators and activating potassium channels, leading to smooth muscle hyperpolarization and relaxation. Depending on VVs severity, management usually includes compression stockings, sclerotherapy and surgical removal. Venotonics have also been promoted to decrease the progression of VVs. Sulodexide has also shown benefits in VLU and CVI, and recent data suggest that it could improve venous smooth muscle contraction. Other lines of treatment including induction of endogenous tissue inhibitors of metalloproteinases (TIMPs) and administration of exogenous synthetic inhibitors of MMPs are being explored, and could provide alternative strategies in the treatment of CVD.
Collapse
Affiliation(s)
- Joseph D Raffetto
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA
| |
Collapse
|
6
|
Rochon ER, Missinato MA, Xue J, Tejero J, Tsang M, Gladwin MT, Corti P. Nitrite Improves Heart Regeneration in Zebrafish. Antioxid Redox Signal 2020; 32:363-377. [PMID: 31724431 PMCID: PMC6985782 DOI: 10.1089/ars.2018.7687] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Aims: Nitrite is reduced to nitric oxide (NO) under physiological and pathological hypoxic conditions to modulate angiogenesis and improve ischemia-reperfusion injury. Although adult mammals lack the ability to regenerate the heart after injury, this is preserved in neonates and efforts to reactivate this process are of great interest. Unlike mammals, the adult zebrafish maintain the innate ability to regenerate their hearts after injury, providing an important model to study cardiac regeneration. We thus explored the effects of physiological levels of nitrite on cardiac and fin regeneration and downstream cellular and molecular signaling pathways in response to amputation and cryoinjury. Results: Nitrite treatment of zebrafish after ventricular amputation or cryoinjury to the heart in hypoxic water (∼3 parts per million of oxygen) increases cardiomyocyte proliferation, improves angiogenesis, and enhances early recruitment of thrombocytes, macrophages, and neutrophils to the injury. When tested in a fin regeneration model, neutrophil recruitment to the injury site was found to be dependent on NO. Innovation: This is the first study to evaluate effects of physiological levels of nitrite on cardiac regeneration in response to cardiac injury, with the observation that nitrite in water accelerates zebrafish heart regeneration. Conclusion: Physiological and therapeutic levels of nitrite increase thrombocyte, neutrophil, and macrophage recruitment to the heart after amputation and cryoinjury in zebrafish, resulting in accelerated cardiomyocyte proliferation and angiogenesis. Translation of this finding to mammalian models of injury during early development may provide an opportunity to improve outcomes during intrauterine fetal or neonatal cardiac surgery.
Collapse
Affiliation(s)
- Elizabeth R Rochon
- Department of Medicine, Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Jianmin Xue
- Department of Medicine, Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jesús Tejero
- Department of Medicine, Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania.,Division of Pulmonary, Department of Medicine, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael Tsang
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- Department of Medicine, Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania.,Division of Pulmonary, Department of Medicine, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Paola Corti
- Department of Medicine, Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania.,Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Ri.MED Foundation, Palermo, Italy
| |
Collapse
|
7
|
Young D, Das N, Anowai A, Dufour A. Matrix Metalloproteases as Influencers of the Cells' Social Media. Int J Mol Sci 2019; 20:E3847. [PMID: 31394726 PMCID: PMC6720954 DOI: 10.3390/ijms20163847] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/01/2019] [Accepted: 08/02/2019] [Indexed: 12/16/2022] Open
Abstract
Matrix metalloproteinases (MMPs) have been studied in the context of cancer due to their ability to increase cell invasion, and were initially thought to facilitate metastasis solely through the degradation of the extracellular matrix (ECM). MMPs have also been investigated in the context of their ECM remodeling activity in several acute and chronic inflammatory diseases. However, after several MMP inhibitors failed in phase III clinical trials, a global reassessment of their biological functions was undertaken, which has revealed multiple unanticipated functions including the processing of chemokines, cytokines, and cell surface receptors. Despite what their name suggests, the matrix aspect of MMPs could contribute to a lesser part of their physiological functions in inflammatory diseases, as originally anticipated. Here, we present examples of MMP substrates implicated in cell signaling, independent of their ECM functions, and discuss the impact for the use of MMP inhibitors.
Collapse
Affiliation(s)
- Daniel Young
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Nabangshu Das
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB T2N 4N1, Canada
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Anthonia Anowai
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Antoine Dufour
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada.
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB T2N 4N1, Canada.
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 4N1, Canada.
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
8
|
Non-Invasive Detection of Extracellular Matrix Metalloproteinase Inducer EMMPRIN, a New Therapeutic Target against Atherosclerosis, Inhibited by Endothelial Nitric Oxide. Int J Mol Sci 2018; 19:ijms19103248. [PMID: 30347750 PMCID: PMC6214015 DOI: 10.3390/ijms19103248] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 09/30/2018] [Accepted: 10/10/2018] [Indexed: 01/12/2023] Open
Abstract
Lack of endothelial nitric oxide causes endothelial dysfunction and circulating monocyte infiltration, contributing to systemic atheroma plaque formation in arterial territories. Among the different inflammatory products, macrophage-derived foam cells and smooth muscle cells synthesize matrix metalloproteinases (MMPs), playing a pivotal role in early plaque formation and enlargement. We found increased levels of MMP-9 and MMP-13 in human endarterectomies with advanced atherosclerosis, together with significant amounts of extracellular matrix (ECM) metalloproteinase inducer EMMPRIN. To test whether the absence of NO may aggravate atherosclerosis through EMMPRIN activation, double NOS3/apoE knockout (KO) mice expressed high levels of EMMPRIN in carotid plaques, suggesting that targeting extracellular matrix degradation may represent a new mechanism by which endothelial NO prevents atherosclerosis. Based on our previous experience, by using gadolinium-enriched paramagnetic fluorescence micellar nanoparticles conjugated with AP9 (NAP9), an EMMPRIN-specific binding peptide, magnetic resonance sequences allowed non-invasive visualization of carotid EMMPRIN in NOS3/apoE over apoE control mice, in which atheroma plaques were significantly reduced. Taken together, these results point to EMMPRIN as a new therapeutic target of NO-mediated protection against atherosclerosis, and NAP9 as a non-invasive molecular tool to target atherosclerosis.
Collapse
|
9
|
Gao J, Jiang L, Liang Q, Shi J, Hou D, Tang D, Chen S, Kong D, Wang S. The grafts modified by heparinization and catalytic nitric oxide generation used for vascular implantation in rats. Regen Biomater 2018; 5:105-114. [PMID: 29644092 PMCID: PMC5888227 DOI: 10.1093/rb/rby003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 02/05/2018] [Accepted: 02/09/2018] [Indexed: 01/03/2023] Open
Abstract
Small-diameter (<6 mm) vascular grafts are increasingly needed in peripheral vascular surgery but have few successes because of acute thrombosis, incomplete endothelialization and intimal hyperplasia after implantation. This study used electrospun poly(ε-caprolactone) as the matrix material. Heparin and selenium-containing catalyst-organoselenium modified polyethyleneimine were introduced through layer-by-layer assembly in order to build a vascular graft with in situ nitric oxide (NO) generation. The aim of this study was to explore the application of the graft with improved histocompatibility and biological function for vascular implantation in rats. After implantation in rats, compared to poly(ε-caprolactone), the modified grafts could promote the adhesion and proliferation of endothelial cells, and inhibit the adhesion of smooth muscle cells. The modified grafts remarkably promoted endothelialization, inhibited intimal hyperplasia and increased the ratio of alternatively activated macrophages (M2) to classical activated macrophages (M1). This work constructed a vascular graft with heparinization and catalytic NO generation for improving the vascularization, and accelerating the tissue regeneration by regulating the inflammatory response. The present study indicates that it is a promising method for regulating response and tissue regeneration of small diameter vascular grafts by a novel approach of combining heparinization and catalytic NO generation.
Collapse
Affiliation(s)
- Jingchen Gao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Li Jiang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Qinge Liang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jie Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Ding Hou
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Di Tang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Siyuan Chen
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shufang Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
10
|
Yao L, Bhatta A, Xu Z, Chen J, Toque HA, Chen Y, Xu Y, Bagi Z, Lucas R, Huo Y, Caldwell RB, Caldwell RW. Obesity-induced vascular inflammation involves elevated arginase activity. Am J Physiol Regul Integr Comp Physiol 2017; 313:R560-R571. [PMID: 28835451 PMCID: PMC5792147 DOI: 10.1152/ajpregu.00529.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 08/08/2017] [Accepted: 08/08/2017] [Indexed: 01/29/2023]
Abstract
Obesity-induced vascular dysfunction involves pathological remodeling of the visceral adipose tissue (VAT) and increased inflammation. Our previous studies showed that arginase 1 (A1) in endothelial cells (ECs) is critically involved in obesity-induced vascular dysfunction. We tested the hypothesis that EC-A1 activity also drives obesity-related VAT remodeling and inflammation. Our studies utilized wild-type and EC-A1 knockout (KO) mice made obese by high-fat/high-sucrose (HFHS) diet. HFHS diet induced increases in body weight, fasting blood glucose, and VAT expansion. This was accompanied by increased arginase activity and A1 expression in vascular ECs and increased expression of tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein-1 (MCP-1), interleukin-10 (IL-10), vascular cell adhesion molecule-1 (VCAM-1), and intercellular adhesion molecule-1 (ICAM-1) mRNA and protein in both VAT and ECs. HFHS also markedly increased circulating inflammatory monocytes and VAT infiltration by inflammatory macrophages, while reducing reparative macrophages. Additionally, adipocyte size and fibrosis increased and capillary density decreased in VAT. These effects of HFHS, except for weight gain and hyperglycemia, were prevented or reduced in mice lacking EC-A1 or treated with the arginase inhibitor 2-(S)-amino-6-boronohexanoic acid (ABH). In mouse aortic ECs, exposure to high glucose (25 mM) and Na palmitate (200 μM) reduced nitric oxide production and increased A1, TNF-α, VCAM-1, ICAM-1, and MCP-1 mRNA, and monocyte adhesion. Knockout of EC-A1 or ABH prevented these effects. HFHS diet-induced VAT inflammation is mediated by EC-A1 expression/activity. Limiting arginase activity is a possible therapeutic means of controlling obesity-induced vascular and VAT inflammation.
Collapse
Affiliation(s)
- Lin Yao
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Anil Bhatta
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Zhimin Xu
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Jijun Chen
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Haroldo A Toque
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yongjun Chen
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Yimin Xu
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Zsolt Bagi
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Rudolf Lucas
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yuqing Huo
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
- Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Ruth B Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
- Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia
- Veterans Administration Medical Center, Augusta, Georgia; and
| | - R William Caldwell
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia;
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
11
|
Brown BA, Williams H, George SJ. Evidence for the Involvement of Matrix-Degrading Metalloproteinases (MMPs) in Atherosclerosis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 147:197-237. [PMID: 28413029 DOI: 10.1016/bs.pmbts.2017.01.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Atherosclerosis leads to blockage of arteries, culminating in myocardial infarction, and stroke. The involvement of matrix-degrading metalloproteinases (MMPs) in atherosclerosis is established and many studies have highlighted the importance of various MMPs in this process. MMPs were first implicated in atherosclerosis due to their ability to degrade extracellular matrix components, which can lead to increased plaque instability. However, more recent work has highlighted a multitude of roles for MMPs in addition to breakdown of extracellular matrix proteins. MMPs are now known to be involved in various stages of plaque progression: from initial macrophage infiltration to plaque rupture. This chapter summarizes the development and progression of atherosclerotic plaques and the contribution of MMPs. We provide data from human studies showing the effect of MMP polymorphisms and the expression of MMPs in both the atherosclerotic plaque and within plasma. We also discuss work in animal models of atherosclerosis that show the effect of gain or loss of function of MMPs. Together, the data provided from these studies illustrate that MMPs are ideal targets as both biomarkers and potential drug therapies for atherosclerosis.
Collapse
Affiliation(s)
- Bethan A Brown
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Helen Williams
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Sarah J George
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom.
| |
Collapse
|
12
|
Yao M, Wang X, Zhao Y, Wang X, Gao F. Expression of MMPs is dependent on the activity of mitogen-activated protein kinase in chondrosarcoma. Mol Med Rep 2016; 15:915-921. [PMID: 28035378 DOI: 10.3892/mmr.2016.6077] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 11/10/2016] [Indexed: 11/05/2022] Open
Abstract
Matrix metalloproteinases (MMPs) and tissue inhibitors of matrix metalloproteinases (TIMPs) serve an important role in chondrosarcoma. The present study investigated whether the expression of MMPs was dependent on the activity of mitogen-activated protein kinase (MAPK) in chondrosarcoma. Surgical pathological specimens were collected to detect MMP-1, MMP-13, TIMP-1, type II collagen and phosphorylated MAPK levels in normal cartilage, enchondroma and chondrosarcoma tissues. The expression of MMP‑1, MMP‑13, TIMP‑1 and type II collagen was investigated utilizing MAPK inhibitors in chondrosarcoma cells. It was noted that the expression levels of MMP‑1, MMP‑13 and TIMP‑1 were increased in chondrosarcoma with the activity of MAPK. After chondrosarcoma cells were pretreated with MAPK inhibitors, the levels of MMP‑1, MMP‑13 and TIMP‑1 were inhibited. Furthermore, MMP‑1 and MMP‑13 are essential in regulating the degradation of type II collagen and decomposing cartilage matrix major. The high expression levels of MMP‑1 and MMP‑13 in chondrosarcoma expedite the invasion by chondrosarcoma cells and their expression can be depressed by MAPK inhibitors.
Collapse
Affiliation(s)
- Min Yao
- Department of Pathology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Xiaomei Wang
- Department of Pathology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Yufeng Zhao
- Department of Pathology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Xiaomeng Wang
- Department of Pathology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Feng Gao
- Department of Pathology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|
13
|
Cuadrado I, Castejon B, Martin AM, Saura M, Reventun-Torralba P, Zamorano JL, Zaragoza C. Nitric Oxide Induces Cardiac Protection by Preventing Extracellular Matrix Degradation through the Complex Caveolin-3/EMMPRIN in Cardiac Myocytes. PLoS One 2016; 11:e0162912. [PMID: 27649573 PMCID: PMC5029905 DOI: 10.1371/journal.pone.0162912] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 08/30/2016] [Indexed: 12/12/2022] Open
Abstract
Inhibition of Extracellular Matrix degradation by nitric oxide (NO) induces cardiac protection against coronary ischemia/reperfusion (IR). Glycosylation of Extracellular Matrix Metalloproteinase Inducer (EMMPRIN) stimulates enzymatic activation of matrix metalloproteinases (MMPs) in the heart, although the mechanisms leading to EMMPRIN glycosylation are poorly understood. We sought to determine if NO may induce cardiac protection by preventing glycosylation of EMMPRIN in a mouse model of IR. Here we found that Caveolin-3 binds to low glycosylated EMMPRIN (LG-EMMPRIN) in cardiac cells and in the hearts of healthy mice, whereas IR disrupted the complex in nitric oxide synthase 2 (NOS2) knockout (KO) mice. By contrast, the binding was partially restored when mice were fed with an NO donor (DEA-NO) in the drinking water, showing a significant reduction on infarct size (NOS2KO: 34.6±5 vs NOS2KO+DEA-NO: 20.7±9), in expression of matrix metalloproteinases, and cardiac performance was improved (left ventricular ejection fraction (LVEF). NOS2KO: 31±4 vs NOS2KO+DEA-NO: 46±6). The role of Caveolin-3/EMMPRIN in NO-mediated cardiac protection was further assayed in Caveolin-3 KO mice, showing no significant improvement on infarct size (Caveolin-3 KO: 34.8±3 vs Caveolin-3 KO+DEA-NO:33.7±5), or in the expression of MMPs, suggesting that stabilization of the complex Caveolin-3/LG-EMMPRIN may play a significant role in the cardioprotective effect of NO against IR.
Collapse
Affiliation(s)
- Irene Cuadrado
- Department of Systems Biology (Physiology), University of Alcalá, School of Medicine (IRYCIS), Ctra. Madrid Barcelona, Km 3,300, 28875, Alcalá de Henares, Madrid, Spain
| | - Borja Castejon
- Cardiology Department, University Francisco de Vitoria/Hospital Ramón y Cajal Research Unit (IRYCIS), Ctra. Colmenar Viejo, km. 9100, 28034, Madrid, Spain
| | - Ana M. Martin
- Cardiology Department, University Francisco de Vitoria/Hospital Ramón y Cajal Research Unit (IRYCIS), Ctra. Colmenar Viejo, km. 9100, 28034, Madrid, Spain
| | - Marta Saura
- Department of Systems Biology (Physiology), University of Alcalá, School of Medicine (IRYCIS), Ctra. Madrid Barcelona, Km 3,300, 28875, Alcalá de Henares, Madrid, Spain
| | - Paula Reventun-Torralba
- Department of Systems Biology (Physiology), University of Alcalá, School of Medicine (IRYCIS), Ctra. Madrid Barcelona, Km 3,300, 28875, Alcalá de Henares, Madrid, Spain
| | - Jose Luis Zamorano
- Cardiology Department, University Hospital Ramón y Cajal (IRYCIS), Ctra Colmenar Viejo, km. 9100, 28034, Madrid, Spain
| | - Carlos Zaragoza
- Cardiology Department, University Francisco de Vitoria/Hospital Ramón y Cajal Research Unit (IRYCIS), Ctra. Colmenar Viejo, km. 9100, 28034, Madrid, Spain
- * E-mail:
| |
Collapse
|
14
|
Effects and mechanism of Xin Mai Jia in a rabbit model of atherosclerosis. Exp Ther Med 2015; 10:1627-1634. [PMID: 26640529 PMCID: PMC4665141 DOI: 10.3892/etm.2015.2774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 10/24/2014] [Indexed: 11/05/2022] Open
Abstract
The aim of this study was to investigate the protective effects of Xin Mai Jia (XMJ) on atherosclerosis (AS) in rabbits and to explore the underlying mechanisms in order to provide experimental evidence for the clinical application of XMJ. An intraperitoneal injection of vitamin D3, combined with a high-fat diet and sacculus injury, was utilized to establish the AS rabbit model. Following the oral administration of lovastatin, Zhibituo and different dosages of XMJ, respectively, blood was drawn from each rabbit for the detection of blood rheological indicators, such as serum lipids. The pathological changes in the right common carotid artery were observed. Vascular function experiments and the expression detection of common carotid artery-related proteins by immunohistochemistry were conducted. XMJ was observed to decrease the blood lipid levels of the AS rabbits; increase the concentration of high-density lipoprotein and apolipoprotein A; decrease blood viscosity, erythrocyte sedimentation rate and hematocrit; elevate the levels of endothelial nitric oxide synthase (eNOS) and Na+/H+ exchanger 1 in vascular tissues and decrease the levels of angiotensin II receptor, type 1 (AT-1) and endothelin-1 (ET-1). In conclusion, XMJ was shown to lower the blood lipid levels of the experimental AS rabbits, improve the abnormal changes in hemorheology, increase the eNOS content in the vascular tissue, decrease the AT-1 and ET-1 levels and increase the endothelium-dependent vasodilation reaction. XMJ therefore has an anti-AS effect.
Collapse
|
15
|
Yang SW, Lim L, Ju S, Choi DH, Song H. Effects of matrix metalloproteinase 13 on vascular smooth muscle cells migration via Akt–ERK dependent pathway. Tissue Cell 2015; 47:115-21. [DOI: 10.1016/j.tice.2014.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 12/12/2014] [Accepted: 12/13/2014] [Indexed: 02/08/2023]
|
16
|
Cao J, Han Z, Tian L, Chen K, Fan Y, Ye B, Huang W, Wang C, Huang Z. Curcumin inhibits EMMPRIN and MMP-9 expression through AMPK-MAPK and PKC signaling in PMA induced macrophages. J Transl Med 2014; 12:266. [PMID: 25241044 PMCID: PMC4205290 DOI: 10.1186/s12967-014-0266-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 09/16/2014] [Indexed: 02/06/2023] Open
Abstract
In coronary arteries, plaque disruption, the major acute clinical manifestations of atherosclerosis, leads to a subsequent cardiac event, such as acute myocardial infarction (AMI) and unstable angina pectoris (UA). Numerous reports have shown that high expression of MMP-9 (matrix metalloproteinase-9), MMP-13 (matrix metalloproteinase-13) and EMMPRIN (extracellular matrix metalloproteinase induce) in monocyte/macrophage results in the plaque progression and destabilization. Curcumin exerts well-known anti-inflammatory and antioxidant effects and probably has a protective role in the atherosclerosis. The purpose of our study was to investigate the molecular mechanisms by which curcumin affects MMP-9, MMP13 and EMMPRIN in PMA (phorbol 12-myristate 13-acetate) induced macrophages. Human monocytic cells (THP-1 cells) were pretreated with curcumin or compound C for 1 h, and then induced by PMA for 48 h. Total RNA and proteins were collected for real-time PCR and Western blot analysis, respectively. In the present study, the exposure to curcumin resulted in attenuated JNK, p38, and ERK activation and decreased expression of MMP-9, MMP-13 and EMMPRIN in PMA induced macrophages. Moreover, we demonstrated that AMPK (AMP-activated protein kinase) and PKC (Protein Kinase C) was activated by PMA during monocyte/macrophage differentiation. Furthermore, curcumin reversed PMA stimulated PKC activation and suppressed the chronic activation of AMPK, which in turn reduced the expression of MMP-9, MMP-13 and EMMPRIN. Therefore, it is suggested that curcumin by inhibiting AMPK-MAPK (mitogen activated protein kinase) and PKC pathway may led to down-regulated EMMPRIN, MMP-9 and MMP-13 expression in PMA-induced THP-1 cells.
Collapse
|
17
|
Saura M, Marquez S, Reventun P, Olea‐Herrero N, Arenas MI, Moreno‐Gómez‐Toledano R, Gómez‐Parrizas M, Muñóz‐Moreno C, González‐Santander M, Zaragoza C, Bosch RJ. Oral administration of bisphenol A induces high blood pressure through angiotensin II/CaMKII‐dependent uncoupling of eNOS. FASEB J 2014; 28:4719-28. [DOI: 10.1096/fj.14-252460] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Marta Saura
- Laboratory of Pathophysiology of the Vascular WallUniversity of AlcaláAlcalá de HenaresSpain
- Physiology UnitDepartment of Systems BiologyUniversity of AlcaláAlcalá de HenaresSpain
| | - Susana Marquez
- Laboratory of Pathophysiology of the Vascular WallUniversity of AlcaláAlcalá de HenaresSpain
- Physiology UnitDepartment of Systems BiologyUniversity of AlcaláAlcalá de HenaresSpain
| | - Paula Reventun
- Laboratory of Pathophysiology of the Vascular WallUniversity of AlcaláAlcalá de HenaresSpain
- Physiology UnitDepartment of Systems BiologyUniversity of AlcaláAlcalá de HenaresSpain
| | - Nuria Olea‐Herrero
- Physiology UnitDepartment of Systems BiologyUniversity of AlcaláAlcalá de HenaresSpain
- Laboratory of Renal Physiology and Experimental NephrologyUniversity of AlcaláAlcalá de HenaresSpain
| | - María Isabel Arenas
- Laboratory of Renal Physiology and Experimental NephrologyUniversity of AlcaláAlcalá de HenaresSpain
- Cell Biology UnitDepartment of Biomedicine and BiotechnologyUniversity of AlcaláAlcalá de HenaresSpain
| | - Rafael Moreno‐Gómez‐Toledano
- Physiology UnitDepartment of Systems BiologyUniversity of AlcaláAlcalá de HenaresSpain
- Laboratory of Renal Physiology and Experimental NephrologyUniversity of AlcaláAlcalá de HenaresSpain
| | | | - Carmen Muñóz‐Moreno
- Physiology UnitDepartment of Systems BiologyUniversity of AlcaláAlcalá de HenaresSpain
- Laboratory of Renal Physiology and Experimental NephrologyUniversity of AlcaláAlcalá de HenaresSpain
| | - Marta González‐Santander
- Laboratory of Renal Physiology and Experimental NephrologyUniversity of AlcaláAlcalá de HenaresSpain
- Department of Medicine and Medical SpecialtiesUniversity of AlcaláAlcalá de HenaresSpain
| | - Carlos Zaragoza
- National Center for Cardiovascular Research (CNIC)MadridSpain
- Laboratory of Cardiovascular PathophysiologyJoint Translational Research UnitUniversity Francisco de Vitoria School of Medicine and Division of CardiologyUniversity Hospital Ramón y CajalMadridSpain
| | - Ricardo J. Bosch
- Physiology UnitDepartment of Systems BiologyUniversity of AlcaláAlcalá de HenaresSpain
- Laboratory of Renal Physiology and Experimental NephrologyUniversity of AlcaláAlcalá de HenaresSpain
| |
Collapse
|
18
|
Lavin B, Gómez M, Pello OM, Castejon B, Piedras MJ, Saura M, Zaragoza C. Nitric oxide prevents aortic neointimal hyperplasia by controlling macrophage polarization. Arterioscler Thromb Vasc Biol 2014; 34:1739-46. [PMID: 24925976 DOI: 10.1161/atvbaha.114.303866] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Nitric oxide synthase 3 (NOS3) prevents neointima hyperplasia by still unknown mechanisms. To demonstrate the significance of endothelial nitric oxide in the polarization of infiltrated macrophages through the expression of matrix metalloproteinase (MMP)-13 in neointima formation. APPROACH AND RESULTS After aortic endothelial denudation, NOS3 null mice show elevated neointima formation, detecting increased mobilization of LSK (lineage-negative [Lin]-stem-cell antigen 1 [SCA1]+KIT+) progenitor cells, and high ratios of M1 (proinflammatory) to M2 (resolving) macrophages, accompanied by high expression of interleukin-5, interleukin-6, MCP-1 (monocyte chemoattractant protein), VEGF (vascular endothelial growth factor), GM-CSF (granulocyte-macrophage colony stimulating factor), interleukin-1β, and interferon-γ. In conditional c-Myc knockout mice, in which M2 polarization is defective, denuded aortas showed extensive wall thickening as well. Conditioned medium from NOS3-deficient endothelium induced extensive repolarization of M2 macrophages to an M1 phenotype, and vascular smooth muscle cells proliferated and migrated faster in conditioned medium from M1 macrophages. Among the different proteins participating in cell migration, MMP-13 was preferentially expressed by M1 macrophages. M1-mediated vascular smooth muscle cell migration was inhibited when macrophages were isolated from MMP-13-deficient mice, whereas exogenous administration of MMP-13 to vascular smooth muscle cell fully restored migration. Excess vessel wall thickening in mice lacking NOS3 was partially reversed by simultaneous deletion of MMP-13, indicating that NOS3 prevents neointimal hyperplasia by preventing MMP-13 activity. An excess of M1-polarized macrophages that coexpress MMP-13 was also detected in human carotid samples from endarterectomized patients. CONCLUSIONS These findings indicate that at least M1 macrophage-mediated expression of MMP-13 in NOS3 null mice induces neointima formation after vascular injury, suggesting that MMP-13 may represent a new promising target in vascular disease.
Collapse
Affiliation(s)
- Begoña Lavin
- From the Fundación Centro Nacional de Investigaciones Cardiovasculares CNIC Melchor Fernandez Almagro 3, Madrid, Spain (B.L., M.G., O.M.P., B.C., C.Z.); Departmento de Fisiología, Facultad de Medicina, Universidad de Alcala, Ctra Madrid-Barcelona, Alcala de Henares, Spain (M.S.); and Facultad de Medicina, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain (M.J.P., C.Z.)
| | - Monica Gómez
- From the Fundación Centro Nacional de Investigaciones Cardiovasculares CNIC Melchor Fernandez Almagro 3, Madrid, Spain (B.L., M.G., O.M.P., B.C., C.Z.); Departmento de Fisiología, Facultad de Medicina, Universidad de Alcala, Ctra Madrid-Barcelona, Alcala de Henares, Spain (M.S.); and Facultad de Medicina, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain (M.J.P., C.Z.)
| | - Oscar M Pello
- From the Fundación Centro Nacional de Investigaciones Cardiovasculares CNIC Melchor Fernandez Almagro 3, Madrid, Spain (B.L., M.G., O.M.P., B.C., C.Z.); Departmento de Fisiología, Facultad de Medicina, Universidad de Alcala, Ctra Madrid-Barcelona, Alcala de Henares, Spain (M.S.); and Facultad de Medicina, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain (M.J.P., C.Z.)
| | - Borja Castejon
- From the Fundación Centro Nacional de Investigaciones Cardiovasculares CNIC Melchor Fernandez Almagro 3, Madrid, Spain (B.L., M.G., O.M.P., B.C., C.Z.); Departmento de Fisiología, Facultad de Medicina, Universidad de Alcala, Ctra Madrid-Barcelona, Alcala de Henares, Spain (M.S.); and Facultad de Medicina, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain (M.J.P., C.Z.)
| | - Maria J Piedras
- From the Fundación Centro Nacional de Investigaciones Cardiovasculares CNIC Melchor Fernandez Almagro 3, Madrid, Spain (B.L., M.G., O.M.P., B.C., C.Z.); Departmento de Fisiología, Facultad de Medicina, Universidad de Alcala, Ctra Madrid-Barcelona, Alcala de Henares, Spain (M.S.); and Facultad de Medicina, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain (M.J.P., C.Z.)
| | - Marta Saura
- From the Fundación Centro Nacional de Investigaciones Cardiovasculares CNIC Melchor Fernandez Almagro 3, Madrid, Spain (B.L., M.G., O.M.P., B.C., C.Z.); Departmento de Fisiología, Facultad de Medicina, Universidad de Alcala, Ctra Madrid-Barcelona, Alcala de Henares, Spain (M.S.); and Facultad de Medicina, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain (M.J.P., C.Z.)
| | - Carlos Zaragoza
- From the Fundación Centro Nacional de Investigaciones Cardiovasculares CNIC Melchor Fernandez Almagro 3, Madrid, Spain (B.L., M.G., O.M.P., B.C., C.Z.); Departmento de Fisiología, Facultad de Medicina, Universidad de Alcala, Ctra Madrid-Barcelona, Alcala de Henares, Spain (M.S.); and Facultad de Medicina, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain (M.J.P., C.Z.).
| |
Collapse
|
19
|
Quillard T, Araújo HA, Franck G, Tesmenitsky Y, Libby P. Matrix metalloproteinase-13 predominates over matrix metalloproteinase-8 as the functional interstitial collagenase in mouse atheromata. Arterioscler Thromb Vasc Biol 2014; 34:1179-86. [PMID: 24723558 DOI: 10.1161/atvbaha.114.303326] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Substantial evidence implicates interstitial collagenases of the matrix metalloproteinase (MMP) family in plaque rupture and fatal thrombosis. Understanding the compensatory mechanisms that may influence the expression of these enzymes and their functions, therefore, has important clinical implications. This study assessed in mice the relative effect of the 2 principal mouse collagenases on collagen content and other plaque characteristics. APPROACH AND RESULTS Apolipoprotein E-deficient (apoE(-/-)) mice, MMP-13(-/-) apoE(-/-), MMP-8(-/-) apoE(-/-) double knockout mice, and MMP-13(-/-) MMP-8(-/-) apoE(-/-) triple knockout mice consumed a high-cholesterol diet for 10 and 24 weeks. Both double knockout and triple knockout mice showed comparable atherosclerotic lesion formation compared with apoE(-/-) controls. Analysis of aortic root sections indicated that lesions of MMP-8/MMP-13-deficient and MMP-13-deficient mice accumulate more fibrillar collagen than apoE(-/-) controls and MMP-8(-/-) apoE(-/-) double knockout. We further tested the relative effect of MMPs on plaque collagenolysis using in situ zymography. MMP-13 deletion alone abrogated collagenolytic activity in lesions, indicating a predominant role for MMP-13 in this process. MMP-13 and MMP-13/MMP-8 deficiency did not alter macrophage content but associated with reduced accumulation of smooth muscle cells. CONCLUSIONS These results show that among MMP interstitial collagenases in mice, MMP-13 prevails over MMP-8 in collagen degradation in atheromata. These findings provide a rationale for the identification and selective targeting a predominant collagenase for modulating key aspects of plaque structure considered critical in clinical complications, although they do not translate directly to human lesions, which also contain MMP-1.
Collapse
Affiliation(s)
- Thibaut Quillard
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Haniel Alves Araújo
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Gregory Franck
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Yevgenia Tesmenitsky
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Peter Libby
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
20
|
Fügl A, Gasser H, Watzak G, Bucher A, Feierfeil J, Jürgens G, Watzek G, Hallström S, Gruber R. S-nitroso albumin enhances bone formation in a rabbit calvaria model. Int J Oral Maxillofac Surg 2014; 43:381-6. [DOI: 10.1016/j.ijom.2013.06.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 05/08/2013] [Accepted: 06/11/2013] [Indexed: 10/26/2022]
|
21
|
Perry SW, Schueckler JM, Burke K, Arcuri GL, Brown EB. Stromal matrix metalloprotease-13 knockout alters Collagen I structure at the tumor-host interface and increases lung metastasis of C57BL/6 syngeneic E0771 mammary tumor cells. BMC Cancer 2013; 13:411. [PMID: 24010522 PMCID: PMC3766650 DOI: 10.1186/1471-2407-13-411] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 08/28/2013] [Indexed: 12/21/2022] Open
Abstract
Background Matrix metalloproteases and collagen are key participants in breast cancer, but their precise roles in cancer etiology and progression remain unclear. MMP13 helps regulate collagen structure and has been ascribed largely harmful roles in cancer, but some studies demonstrate that MMP13 may also protect against tumor pathology. Other studies indicate that collagen’s organizational patterns at the breast tumor-host interface influence metastatic potential. Therefore we investigated how MMP13 modulates collagen I, a principal collagen subtype in breast tissue, and affects tumor pathology and metastasis in a mouse model of breast cancer. Methods Tumors were implanted into murine mammary tissues, and their growth analyzed in Wildtype and MMP13 KO mice. Following extraction, tumors were analyzed for collagen I levels and collagen I macro- and micro-structural properties at the tumor-host boundary using immunocytochemistry and two-photon and second harmonic generation microscopy. Lungs were analyzed for metastases counts, to correlate collagen I changes with a clinically significant functional parameter. Statistical analyses were performed by t-test, analysis of variance, or Wilcoxon-Mann–Whitney tests as appropriate. Results We found that genetic ablation of host stromal MMP13 led to: 1. Increased mammary tumor collagen I content, 2. Marked changes in collagen I spatial organization, and 3. Altered collagen I microstructure at the tumor-host boundary, as well as 4. Increased metastasis from the primary mammary tumor to lungs. Conclusions These results implicate host MMP13 as a key regulator of collagen I structure and metastasis in mammary tumors, thus making it an attractive potential therapeutic target by which we might alter metastatic potential, one of the chief determinants of clinical outcome in breast cancer. In addition to identifying stromal MMP13 is an important regulator of the tumor microenvironment and metastasis, these results also suggest that stromal MMP13 may protect against breast cancer pathology under some conditions, a finding with important implications for development of chemotherapies directed against matrix metalloproteases.
Collapse
Affiliation(s)
- Seth W Perry
- Department of Biomedical Engineering, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| | | | | | | | | |
Collapse
|
22
|
Xiao X, Mruk DD, Cheng CY. Intercellular adhesion molecules (ICAMs) and spermatogenesis. Hum Reprod Update 2013; 19:167-86. [PMID: 23287428 DOI: 10.1093/humupd/dms049] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND During the seminiferous epithelial cycle, restructuring takes places at the Sertoli-Sertoli and Sertoli-germ cell interface to accommodate spermatogonia/spermatogonial stem cell renewal via mitosis, cell cycle progression and meiosis, spermiogenesis and spermiation since developing germ cells, in particular spermatids, move 'up and down' the seminiferous epithelium. Furthermore, preleptotene spermatocytes differentiated from type B spermatogonia residing at the basal compartment must traverse the blood-testis barrier (BTB) to enter the adluminal compartment to prepare for meiosis at Stage VIII of the epithelial cycle, a process also accompanied by the release of sperm at spermiation. These cellular events that take place at the opposite ends of the epithelium are co-ordinated by a functional axis designated the apical ectoplasmic specialization (ES)-BTB-basement membrane. However, the regulatory molecules that co-ordinate cellular events in this axis are not known. METHODS Literature was searched at http://www.pubmed.org and http://scholar.google.com to identify published findings regarding intercellular adhesion molecules (ICAMs) and the regulation of this axis. RESULTS Members of the ICAM family, namely ICAM-1 and ICAM-2, and the biologically active soluble ICAM-1 (sICAM-1) are the likely regulatory molecules that co-ordinate these events. sICAM-1 and ICAM-1 have antagonistic effects on the Sertoli cell tight junction-permeability barrier, involved in Sertoli cell BTB restructuring, whereas ICAM-2 is restricted to the apical ES, regulating spermatid adhesion during the epithelial cycle. Studies in other epithelia/endothelia on the role of the ICAM family in regulating cell movement are discussed and this information has been evaluated and integrated into studies of these proteins in the testis to create a hypothetical model, depicting how ICAMs regulate junction restructuring events during spermatogenesis. CONCLUSIONS ICAMs are crucial regulatory molecules of spermatogenesis. The proposed hypothetical model serves as a framework in designing functional experiments for future studies.
Collapse
Affiliation(s)
- Xiang Xiao
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, USA
| | | | | |
Collapse
|
23
|
Tan RJ, Liu Y. Matrix metalloproteinases in kidney homeostasis and diseases. Am J Physiol Renal Physiol 2012; 302:F1351-61. [PMID: 22492945 DOI: 10.1152/ajprenal.00037.2012] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases that have been increasingly linked to both normal physiology and abnormal pathology in the kidney. Collectively able to degrade all components of the extracellular matrix, MMPs were originally thought to antagonize the development of fibrotic diseases solely through digestion of excessive matrix. However, increasing evidence has shown that MMPs play a wide variety of roles in regulating inflammation, epithelial-mesenchymal transition, cell proliferation, angiogenesis, and apoptosis. We now have robust evidence for MMP dysregulation in a multitude of renal diseases including acute kidney injury, diabetic nephropathy, glomerulonephritis, inherited kidney disease, and chronic allograft nephropathy. The goal of this review is to summarize current findings regarding the role of MMPs in kidney diseases as well as the mechanisms of action of this family of proteases.
Collapse
Affiliation(s)
- Roderick J Tan
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, 200 Lothrop St., Pittsburgh, PA 15261, USA
| | | |
Collapse
|
24
|
Suckling K. The landscape of drug discovery in atherosclerosis and dyslipidaemia revisited: an update of patenting activity. Expert Opin Ther Pat 2012; 22:199-204. [PMID: 22404075 DOI: 10.1517/13543776.2012.667402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
INTRODUCTION This paper is an update of a previous paper I published in Expert Opinion of Therapeutic Patents in 2008. The paper was a survey of patenting activity in the fields of atherosclerosis and dyslipidaemia, which identified trends in patenting by reviewing two major mechanistic categories, metabolic/dyslipidaemia and vascular/inflammation, as well as examining the interest in certain specific targets over a period of 10 years. METHODS In this update, the same methodology was followed using the Espacenet of the European Patent Office (EPO) to identify patents claiming therapeutics for atherosclerosis or dyslipidaemia (excluding the wider metabolic syndrome). EXPERT OPINION A major change in the field over the past 5 years has been the departure of larger companies from the field. This is reflected in the patenting activity. Patenting has been at a stable rate over the recent years with few new targets being highlighted. It is suggested that, for this field to return to the higher rates of patenting seen over 10 years ago, breakthroughs in translational medicine and in the ability to conduct clinical trials, particularly in biomarkers and imaging, will need to take place.
Collapse
Affiliation(s)
- Keith Suckling
- Suckling Science Ltd, 291 Knightsfield, Welwyn Garden City, Herts, AL8 7NH, UK.
| |
Collapse
|
25
|
Hemodialysis removes uremic toxins that alter the biological actions of endothelial cells. PLoS One 2012; 7:e30975. [PMID: 22383985 PMCID: PMC3284471 DOI: 10.1371/journal.pone.0030975] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 12/29/2011] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease is linked to systemic inflammation and to an increased risk of ischemic heart disease and atherosclerosis. Endothelial dysfunction associates with hypertension and vascular disease in the presence of chronic kidney disease but the mechanisms that regulate the activation of the endothelium at the early stages of the disease, before systemic inflammation is established remain obscure. In the present study we investigated the effect of serum derived from patients with chronic kidney disease either before or after hemodialysis on the activation of human endothelial cells in vitro, as an attempt to define the overall effect of uremic toxins at the early stages of endothelial dysfunction. Our results argue that uremic toxins alter the biological actions of endothelial cells and the remodelling of the extracellular matrix before signs of systemic inflammatory responses are observed. This study further elucidates the early events of endothelial dysfunction during toxic uremia conditions allowing more complete understanding of the molecular events as well as their sequence during progressive renal failure.
Collapse
|
26
|
Quillard T, Tesmenitsky Y, Croce K, Travers R, Shvartz E, Koskinas KC, Sukhova GK, Aikawa E, Aikawa M, Libby P. Selective inhibition of matrix metalloproteinase-13 increases collagen content of established mouse atherosclerosis. Arterioscler Thromb Vasc Biol 2012; 31:2464-72. [PMID: 21903941 DOI: 10.1161/atvbaha.111.231563] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Evidence has linked collagen loss with the onset of acute coronary events. This study tested the hypothesis that selective matrix metalloproteinase-13 (MMP-13) collagenase inhibition increases collagen content in already established and nascent mouse atheromas. METHODS AND RESULTS In vitro and in situ experiments documented the selectivity and efficacy of an orally available MMP-13 inhibitor (MMP13i-A). In vivo observations monitored macrophage accumulation and MMP-13 activity using molecular imaging. After 10 weeks of MMP13i-A treatment, apolipoprotein E-deficient mice with evolving or established lesions exhibited reduced MMP-13 activity without affecting macrophage content, measured either by intravital microscopy or fluorescence reflectance imaging. Histological analysis indicated that MMP13-iA did not affect plaque size or macrophage or smooth muscle cell accumulation. Administration of MMP13i-A to mice with evolving or established atheromas substantially increased plaque interstitial collagen content in the intima and locally in the fibrous cap, compared with vehicle-treated controls. Analysis of collagen revealed thicker collagen fibers within the plaques of treated groups. CONCLUSION Pharmacological MMP-13 inhibition yields collagen accumulation in plaques (a feature associated in humans with resistance to rupture), even in established plaques. This study, of considerable clinical relevance, furnishes new mechanistic insight into regulation of the plaque's extracellular matrix and validates molecular imaging for studying plaque biology.
Collapse
Affiliation(s)
- Thibaut Quillard
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Matrix metalloproteinase inhibitors as investigative tools in the pathogenesis and management of vascular disease. EXPERIENTIA SUPPLEMENTUM (2012) 2012; 103:209-79. [PMID: 22642194 DOI: 10.1007/978-3-0348-0364-9_7] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Matrix metalloproteinases (MMPs) are proteolytic enzymes that degrade various components of the extracellular matrix (ECM). MMPs could also regulate the activity of several non-ECM bioactive substrates and consequently affect different cellular functions. Members of the MMPs family include collagenases, gelatinases, stromelysins, matrilysins, membrane-type MMPs, and others. Pro-MMPs are cleaved into active MMPs, which in turn act on various substrates in the ECM and on the cell surface. MMPs play an important role in the regulation of numerous physiological processes including vascular remodeling and angiogenesis. MMPs may also be involved in vascular diseases such as hypertension, atherosclerosis, aortic aneurysm, and varicose veins. MMPs also play a role in the hemodynamic and vascular changes associated with pregnancy and preeclampsia. The role of MMPs is commonly assessed by measuring their gene expression, protein amount, and proteolytic activity using gel zymography. Because there are no specific activators of MMPs, MMP inhibitors are often used to investigate the role of MMPs in different physiologic processes and in the pathogenesis of specific diseases. MMP inhibitors include endogenous tissue inhibitors (TIMPs) and pharmacological inhibitors such as zinc chelators, doxycycline, and marimastat. MMP inhibitors have been evaluated as diagnostic and therapeutic tools in cancer, autoimmune disease, and cardiovascular disease. Although several MMP inhibitors have been synthesized and tested both experimentally and clinically, only one MMP inhibitor, i.e., doxycycline, is currently approved by the Food and Drug Administration. This is mainly due to the undesirable side effects of MMP inhibitors especially on the musculoskeletal system. While most experimental and clinical trials of MMP inhibitors have not demonstrated significant benefits, some trials still showed promising results. With the advent of new genetic and pharmacological tools, disease-specific MMP inhibitors with fewer undesirable effects are being developed and could be useful in the management of vascular disease.
Collapse
|
28
|
Lee S, Eskin SG, Shah AK, Schildmeyer LA, McIntire LV. Effect of zinc and nitric oxide on monocyte adhesion to endothelial cells under shear stress. Ann Biomed Eng 2011; 40:697-706. [PMID: 22009315 DOI: 10.1007/s10439-011-0434-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 10/06/2011] [Indexed: 11/26/2022]
Abstract
This study describes the effect of zinc on monocyte adhesion to endothelial cells under different shear stress regimens, which may trigger atherogenesis. Human umbilical vein endothelial cells were exposed to steady shear stress (15 dynes/cm(2) or 1 dyne/cm(2)) or reversing shear stress (time average 1 dyne/cm(2)) for 24 h. In all shear stress regimes, zinc deficiency enhanced THP-1 cell adhesion, while heparinase III reduced monocyte adhesion following reversing shear stress exposure. Unlike other shear stress regimes, reversing shear stress alone enhanced monocyte adhesion, which may be associated with increased H(2)O(2) and superoxide together with relatively low levels of nitric oxide (NO) production. L-N(G)-Nitroarginine methyl ester (L-NAME) treatment increased monocyte adhesion under 15 dynes/cm(2) and under reversing shear stress. After reversing shear stress, monocyte adhesion dramatically increased with heparinase III treatment followed by a zinc scavenger. Static culture experiments supported the reduction of monocyte adhesion by zinc following endothelial cell cytokine activation. These results suggest that endothelial cell zinc levels are important for the inhibition of monocyte adhesion to endothelial cells, and may be one of the key factors in the early stages of atherogenesis.
Collapse
Affiliation(s)
- Sungmun Lee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University School of Medicine, Atlanta, GA 30332-0535, USA
| | | | | | | | | |
Collapse
|
29
|
Morrison C, Mancini S, Cipollone J, Kappelhoff R, Roskelley C, Overall C. Microarray and proteomic analysis of breast cancer cell and osteoblast co-cultures: role of osteoblast matrix metalloproteinase (MMP)-13 in bone metastasis. J Biol Chem 2011; 286:34271-85. [PMID: 21784845 PMCID: PMC3190775 DOI: 10.1074/jbc.m111.222513] [Citation(s) in RCA: 51] [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: 01/17/2011] [Revised: 07/20/2011] [Indexed: 12/20/2022] Open
Abstract
Dynamic reciprocal interactions between a tumor and its microenvironment impact both the establishment and progression of metastases. These interactions are mediated, in part, through proteolytic sculpting of the microenvironment, particularly by the matrix metalloproteinases, with both tumors and stroma contributing to the proteolytic milieu. Because bone is one of the predominant sites of breast cancer metastases, we used a co-culture system in which a subpopulation of the highly invasive human breast cancer cell line MDA-MB-231, with increased propensity to metastasize to bone, was overlaid onto a monolayer of differentiated osteoblast MC3T3-E1 cells in a mineralized osteoid matrix. CLIP-CHIP® microarrays identified changes in the complete protease and inhibitor expression profile of the breast cancer and osteoblast cells that were induced upon co-culture. A large increase in osteoblast-derived MMP-13 mRNA and protein was observed. Affymetrix analysis and validation showed induction of MMP-13 was initiated by soluble factors produced by the breast tumor cells, including oncostatin M and the acute response apolipoprotein SAA3. Significant changes in the osteoblast secretomes upon addition of MMP-13 were identified by degradomics from which six novel MMP-13 substrates with the potential to functionally impact breast cancer metastasis to bone were identified and validated. These included inactivation of the chemokines CCL2 and CCL7, activation of platelet-derived growth factor-C, and cleavage of SAA3, osteoprotegerin, CutA, and antithrombin III. Hence, the influence of breast cancer metastases on the bone microenvironment that is executed via the induction of osteoblast MMP-13 with the potential to enhance metastases growth by generating a microenvironmental amplifying feedback loop is revealed.
Collapse
Affiliation(s)
- Charlotte Morrison
- From the Centre for Blood Research and
- Departments of Oral Biological and Medical Sciences
| | - Stephanie Mancini
- Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Jane Cipollone
- Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Reinhild Kappelhoff
- From the Centre for Blood Research and
- Departments of Oral Biological and Medical Sciences
| | - Calvin Roskelley
- Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Christopher Overall
- From the Centre for Blood Research and
- Departments of Oral Biological and Medical Sciences
- Biochemistry and Molecular Biology, and
| |
Collapse
|
30
|
Tarin C, Lavin B, Gomez M, Saura M, Diez-Juan A, Zaragoza C. The extracellular matrix metalloproteinase inducer EMMPRIN is a target of nitric oxide in myocardial ischemia/reperfusion. Free Radic Biol Med 2011; 51:387-95. [PMID: 21570464 DOI: 10.1016/j.freeradbiomed.2011.04.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 03/29/2011] [Accepted: 04/11/2011] [Indexed: 12/20/2022]
Abstract
Nitric oxide (NO) is an important defense against myocardial ischemia/reperfusion (I/R) injury. Although matrix metalloproteinase (MMP)-mediated necrosis of cardiac myocytes is well characterized, the role of inducible NO synthase (iNOS)-derived NO in this process is poorly understood. I/R injury was increased in iNOS-deficient mice and in mice treated with 1400 W (a pharmacological iNOS inhibitor) and was associated with significantly increased expression of extracellular matrix metalloproteinase inducer (EMMPRIN) and EMMPRIN-associated MMPs. Transcriptional activity of an EMMPRIN luciferase promoter reporter expressed in cardiac myocytes was inhibited by NO in a cGMP-dependent manner, and this transcriptional inhibition was abolished by mutation of a putative E2F site. Consistent with these findings, EMMPRIN null mice, in which iNOS is normally induced, are partially protected against I/R injury. Pharmacological inhibition of iNOS in EMMPRIN null mice had no additional protective effect, suggesting that EMMPRIN is a downstream target of NO. Administration of anti-EMMPRIN neutralizing antibodies partly reduced the excess heart damage and MMP-9 expression induced by I/R in iNOS null mice, indicating that regulation of EMMPRIN is an important mechanism of NO-mediated cardioprotection.
Collapse
Affiliation(s)
- Carlos Tarin
- Fundación Centro Nacional de Investigaciones Cardiovasculares, Madrid 28029, Spain
| | | | | | | | | | | |
Collapse
|
31
|
Li R, Lau WB, Ma XL. Adiponectin resistance and vascular dysfunction in the hyperlipidemic state. Acta Pharmacol Sin 2010; 31:1258-66. [PMID: 20802503 PMCID: PMC4012912 DOI: 10.1038/aps.2010.95] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Accepted: 06/22/2010] [Indexed: 12/16/2022] Open
Abstract
Insulin plays an important role in the stimulation of vascular nitric oxide production, with both short term (vasomotility and anti-thrombotic effects) and long term (smooth muscle cell growth and migration inhibition) benefits. Impaired vasodilatory response to insulin, the hallmark of vascular insulin resistance (IR), has important implications for circulatory pathophysiology. An association between adipokines and IR has been observed in both diabetic and nondiabetic states. Adiponectin (APN) is an insulin-sensitizing adipokine known to stimulate skeletal muscle fatty acid (FA) oxidation and reduce lipid accumulation. Recent demonstrations of potential cross-talk between APN and insulin in vascular function regulation are particularly interesting. The lipid accumulation observed after chronic high-fat (HF) diets and in the obese state may reduce vascular response to APN, a pathologic state termed as APN resistance. This review highlights the importance of insulin sensitivity and APN activity in the maintenance of endothelial function. It explores the relationships between vascular IR and APN resistance in the hyperlipidemic pathological condition, representative of the metabolic syndrome. The investigation of vascular insulin and APN resistance provides not only better understanding of vascular pathophysiology, but also an opportunity for therapeutic targeting in individuals affected by the metabolic syndrome.
Collapse
Affiliation(s)
- Rong Li
- Department of Geriatrics, Xijing Hospital, Xi-an 710032, China
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Xin Liang Ma
- Department of Geriatrics, Xijing Hospital, Xi-an 710032, China
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
32
|
Meierjohann S, Hufnagel A, Wende E, Kleinschmidt MA, Wolf K, Friedl P, Gaubatz S, Schartl M. MMP13 mediates cell cycle progression in melanocytes and melanoma cells: in vitro studies of migration and proliferation. Mol Cancer 2010; 9:201. [PMID: 20667128 PMCID: PMC2915980 DOI: 10.1186/1476-4598-9-201] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Accepted: 07/28/2010] [Indexed: 11/10/2022] Open
Abstract
Background Melanoma cells are usually characterized by a strong proliferative potential and efficient invasive migration. Among the multiple molecular changes that are recorded during progression of this disease, aberrant activation of receptor tyrosine kinases (RTK) is often observed. Activation of matrix metalloproteases goes along with RTK activation and usually enhances RTK-driven migration. The purpose of this study was to examine RTK-driven three-dimensional migration of melanocytes and the pro-tumorigenic role of matrix metalloproteases for melanocytes and melanoma cells. Results Using experimental melanocyte dedifferentiation as a model for early melanomagenesis we show that an activated EGF receptor variant potentiates migration through three-dimensional fibrillar collagen. EGFR stimulation also resulted in a strong induction of matrix metalloproteases in a MAPK-dependent manner. However, neither MAPK nor MMP activity were required for migration, as the cells migrated in an entirely amoeboid mode. Instead, MMPs fulfilled a function in cell cycle regulation, as their inhibition resulted in strong growth inhibition of melanocytes. The same effect was observed in the human melanoma cell line A375 after stimulation with FCS. Using sh- and siRNA techniques, we could show that MMP13 is the protease responsible for this effect. Along with decreased proliferation, knockdown of MMP13 strongly enhanced pigmentation of melanocytes. Conclusions Our data show for the first time that growth stimuli are mediated via MMP13 in melanocytes and melanoma, suggesting an autocrine MMP13-driven loop. Given that MMP13-specific inhibitors are already developed, these results support the evaluation of these inhibitors in the treatment of melanoma.
Collapse
Affiliation(s)
- Svenja Meierjohann
- Department of Physiological Chemistry I, Biocenter, University of Wurzburg, Am Hubland, 97074 Wuerzburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Rainwater DL, Shi Q, Mahaney MC, Hodara V, Vandeberg JL, Wang XL. Genetic regulation of endothelial inflammatory responses in baboons. Arterioscler Thromb Vasc Biol 2010; 30:1628-33. [PMID: 20508207 DOI: 10.1161/atvbaha.110.205740] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To investigate the genetic contributions to the expression of cell surface adhesion molecules on endothelial cells (ECs) and to the release by ECs of chemokines, which are responsible for local inflammation. METHODS AND RESULTS Monocyte adhesion to ECs and transmigration across the endothelial barrier are the key steps in the formation of atherosclerotic plaques and the rupture of the existing plaques. Biopsy specimens were obtained from the femoral arteries of 131 pedigreed baboons (65 males and 66 females) aged 10.4+/-1.5 years (mean+/-SD); arterial ECs were harvested and cultured up to the second passage and then subjected to in vitro challenge with tumor necrosis factor (TNF) alpha, 10 ng/mL, or vehicle for 4 hours. Endothelial surface adhesion molecules were measured using flow cytometry, and chemokines released by the ECs were measured by immunoassay. In response to TNF-alpha treatment, interleukin 8 and monocyte chemoattractant protein-1 released by ECs were increased 3.4- and 26-fold, respectively (P<0.001). The expressions of E-selectin, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1 were increased 12.2-, 41.4-, and 3.5-fold, respectively (P<0.001). The quantitative levels of several traits were heritable after TNF-alpha stimulation: h(2)=0.24 (P=0.02) for interleukin 8 and h(2)=0.28 (P=0.003) for E-selectin in culture medium; h(2)=0.21 (P=0.03) for intercellular adhesion molecule-1; and h(2)=0.37 (P<0.001) for vascular cell adhesion molecule-1 expression on EC surfaces. Furthermore, significant heritability was observed for lysate protein level, which is a measure of cell growth rate, with (h(2)=0.64, P<0.001) or without (h(2)=0.51, P<0.001) TNF-alpha stimulation. CONCLUSIONS This study reports on the heritability of adhesion molecules in ECs when activated by TNF-alpha. This finding suggests genetic regulation of key arterial wall inflammatory processes that are responsible for the initiation of atherosclerotic lesions and the plaque rupture of existing atheromas.
Collapse
Affiliation(s)
- David L Rainwater
- Department of Genetics, Southwest Foundation for Biomedical Research, San Antonio, TX, USA
| | | | | | | | | | | |
Collapse
|
34
|
Parkington HC, Coleman HA, Wintour EM, Tare M. Prenatal alcohol exposure: implications for cardiovascular function in the fetus and beyond. Clin Exp Pharmacol Physiol 2009; 37:e91-8. [PMID: 19930419 DOI: 10.1111/j.1440-1681.2009.05342.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
1. The effects of heavy maternal alcohol consumption during pregnancy on cognitive and behavioural performance and craniofacial malformations in the offspring have been studied extensively. In contrast, the impact of maternal alcohol intake on the cardiovascular system of the offspring and the effects of more modest consumption have received very scant consideration. 2. Adverse conditions in the pre- and neonatal periods can have a profound legacy on offspring health, including the risk of cardiovascular disease. Prenatal alcohol exposure can modulate vascular reactivity, including endothelial and smooth muscle function. 3. Other effects of prenatal alcohol exposure are emerging, including impairment of nephrogenesis and kidney function and increased arterial stiffness. The impact of even modest prenatal alcohol exposure on cardiovascular health in the offspring remains to be determined. 4. It is envisaged that the culmination of reduced renal and vascular capacity will render the offspring more vulnerable to cardiovascular disease with ageing and exposure to additional insults and lifestyle factors.
Collapse
Affiliation(s)
- Helena C Parkington
- Department of Physiology, Monash University, Melbourne, Victoria, Australia.
| | | | | | | |
Collapse
|
35
|
Lizarbe TR, Tarín C, Gómez M, Lavin B, Aracil E, Orte LM, Zaragoza C. Nitric oxide induces the progression of abdominal aortic aneurysms through the matrix metalloproteinase inducer EMMPRIN. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:1421-30. [PMID: 19779140 DOI: 10.2353/ajpath.2009.080845] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Nitric Oxide (NO) is involved in the development and progression of abdominal aortic aneurysms (AAA). We found that inhibition of inducible NO synthase (iNOS) protects mice in an elastase-induced AAA model, significantly inhibiting the production of matrix metalloproteinase-13 (MMP-13). The extracellular MMP inducer (EMMPRIN; CD147) was increased in human AAA biopsies and in wild-type murine AAA but not in AAA from iNOS null mice. In cells overexpressing ectopic EMMPRIN, MMP-13 secretion was stimulated, whereas silencing of EMMPRIN by RNA interference led to significant inhibition of MMP-13 expression. In addition, elastase infusion of MMP-13 null mouse aortas induced a significant increase of EMMPRIN but reduced aortic dilatation when compared with wild-type mice, suggesting that NO-mediated AAA may be mediated through EMMPRIN induction of MMP-13. These findings were further verified in elastase-infused iNOS null mice, in which daily administration of NO caused a significant aortic dilatation and the expression of EMMPRIN and MMP-13. By contrast, in iNOS wild-type mice, pharmacological inhibition of iNOS by administration of 1400 W induced a reduction of aortic diameter and inhibition of MMP-13 and EMMPRIN expression when compared with control mice. Our results suggest that NO may regulate the development of AAA in part by inducing the expression of EMMPRIN and modulating the activity of MMP-13 in murine and human aneurysms.
Collapse
Affiliation(s)
- Tania R Lizarbe
- Institutional Fundación Centro Nacional de Investigaciones Cardiovasculares, Madrid 28029, Spain
| | | | | | | | | | | | | |
Collapse
|
36
|
The expression of matrix metalloproteinase-13 is increased in vessels with blood–brain barrier impairment in a stroke-prone hypertensive model. Hypertens Res 2009; 32:332-8. [DOI: 10.1038/hr.2009.26] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|