Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Nature ; 626(8001): 1108-1115, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38326622

ABSTRACT

Psychosocial stress has profound effects on the body, including the immune system and the brain1,2. Although a large number of pre-clinical and clinical studies have linked peripheral immune system alterations to stress-related disorders such as major depressive disorder (MDD)3, the underlying mechanisms are not well understood. Here we show that expression of a circulating myeloid cell-specific proteinase, matrix metalloproteinase 8 (MMP8), is increased in the serum of humans with MDD as well as in stress-susceptible mice following chronic social defeat stress (CSDS). In mice, we show that this increase leads to alterations in extracellular space and neurophysiological changes in the nucleus accumbens (NAc), as well as altered social behaviour. Using a combination of mass cytometry and single-cell RNA sequencing, we performed high-dimensional phenotyping of immune cells in circulation and in the brain and demonstrate that peripheral monocytes are strongly affected by stress. In stress-susceptible mice, both circulating monocytes and monocytes that traffic to the brain showed increased Mmp8 expression following chronic social defeat stress. We further demonstrate that circulating MMP8 directly infiltrates the NAc parenchyma and controls the ultrastructure of the extracellular space. Depleting MMP8 prevented stress-induced social avoidance behaviour and alterations in NAc neurophysiology and extracellular space. Collectively, these data establish a mechanism by which peripheral immune factors can affect central nervous system function and behaviour in the context of stress. Targeting specific peripheral immune cell-derived matrix metalloproteinases could constitute novel therapeutic targets for stress-related neuropsychiatric disorders.


Subject(s)
Depressive Disorder, Major , Matrix Metalloproteinase 8 , Monocytes , Stress, Psychological , Animals , Humans , Mice , Depressive Disorder, Major/blood , Depressive Disorder, Major/enzymology , Depressive Disorder, Major/genetics , Depressive Disorder, Major/metabolism , Extracellular Space/metabolism , Matrix Metalloproteinase 8/blood , Matrix Metalloproteinase 8/deficiency , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase 8/metabolism , Mice, Inbred C57BL , Monocytes/chemistry , Monocytes/immunology , Monocytes/metabolism , Nucleus Accumbens/metabolism , Nucleus Accumbens/pathology , Parenchymal Tissue/metabolism , Single-Cell Gene Expression Analysis , Social Behavior , Social Isolation , Stress, Psychological/blood , Stress, Psychological/genetics , Stress, Psychological/immunology , Stress, Psychological/metabolism
2.
Am J Respir Crit Care Med ; 204(12): 1433-1451, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34550870

ABSTRACT

Rationale: Mechanical signaling through cell-matrix interactions plays a major role in progressive vascular remodeling in pulmonary arterial hypertension (PAH). MMP-8 (matrix metalloproteinase-8) is an interstitial collagenase involved in regulating inflammation and fibrosis of the lung and systemic vasculature, but its role in PAH pathogenesis remains unexplored. Objectives: To evaluate MMP-8 as a modulator of pathogenic mechanical signaling in PAH. Methods: MMP-8 levels were measured in plasma from patients with pulmonary hypertension (PH) and controls by ELISA. MMP-8 vascular expression was examined in lung tissue from patients with PAH and rodent models of PH. MMP-8-/- and MMP-8+/+ mice were exposed to normobaric hypoxia or normoxia for 4-8 weeks. PH severity was evaluated by right ventricular systolic pressure, echocardiography, pulmonary artery morphometry, and immunostaining. Proliferation, migration, matrix component expression, and mechanical signaling were assessed in MMP-8-/- and MMP-8+/+ pulmonary artery smooth muscle cells (PASMCs). Measurements and Main Results: MMP-8 expression was significantly increased in plasma and pulmonary arteries of patients with PH compared with controls and induced in the pulmonary vasculature in rodent PH models. Hypoxia-exposed MMP-8-/- mice had significant mortality, increased right ventricular systolic pressure, severe right ventricular dysfunction, and exaggerated vascular remodeling compared with MMP-8+/+ mice. MMP-8-/- PASMCs demonstrated exaggerated proliferation and migration mediated by altered matrix protein expression, elevated integrin-ß3 levels, and induction of FAK (focal adhesion kinase) and downstream YAP (Yes-associated protein)/TAZ (transcriptional coactivator with PDZ-binding motif) activity. Conclusions: MMP-8 is a novel protective factor upregulated in the pulmonary vasculature during PAH pathogenesis. MMP-8 opposes pathologic mechanobiological feedback by altering matrix composition and disrupting integrin-ß3/FAK and YAP/TAZ-dependent mechanical signaling in PASMCs.


Subject(s)
Matrix Metalloproteinase 8/metabolism , Pulmonary Arterial Hypertension/metabolism , Pulmonary Artery/metabolism , Adult , Aged , Animals , Biomarkers/metabolism , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Matrix Metalloproteinase 8/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Pulmonary Arterial Hypertension/pathology , Pulmonary Arterial Hypertension/prevention & control , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley , Up-Regulation , Vascular Remodeling
3.
Cardiovasc Res ; 116(1): 211-225, 2020 01 01.
Article in English | MEDLINE | ID: mdl-30778537

ABSTRACT

AIMS: Emerging evidence has suggested that adventitia stem/progenitor cells (AdSPCs) migrate into the intima of arteries in response to injury, where they differentiate towards smooth muscle cells (SMCs) and participate in neointimal hyperplasia. We have previously identified matrix metalloproteinase-8 (MMP8) as a key player in atherogenesis. In this study, we aimed to investigate the functional roles of macrophage-derived MMP8 in AdSPC differentiation and injury-induced arterial remodelling. METHODS AND RESULTS: We first observed an important role for MMP8 in SMC differentiation from embryonic stem cells, but this effect was not seen in AdSPCs. Instead, through macrophages/AdSPCs co-culture and macrophage conditional culture medium studies, we have demonstrated that the MMP8 protein secreted from macrophages promotes SMC differentiation from AdSPCs. Mechanistically, we showed that macrophage-derived MMP8 promotes SMC differentiation from AdSPCs through modulating transforming growth factor-ß activity and a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10)/Notch1 signalling. We further demonstrated that the binding site for CBF1, Suppressor of Hairless, and Lag-1 (CSL) within SMC gene promoters is responsible for Notch1 mediated SMC differentiation. Finally, we demonstrated that macrophage-derived MMP8 increased injury-induced neointimal SMC hyperplasia by activating ADAM10/Notch1 signalling. CONCLUSIONS: We have identified macrophage-derived MMP8 as a regulator in SMC differentiation from AdSPCs and neointimal SMC hyperplasia in response to injury. Our data provide new insights into the roles of MMP8 in AdSPC differentiation and the pathogenesis of neointima formation in the context of angiographic restenosis, and therefore may aid in the development of novel therapeutic agents for the prevention of this disease.


Subject(s)
Adventitia/enzymology , Carotid Artery Injuries/enzymology , Cell Differentiation , Cell Proliferation , Macrophages/enzymology , Matrix Metalloproteinase 8/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Neointima , Stem Cells/enzymology , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Adventitia/pathology , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Macrophages/pathology , Matrix Metalloproteinase 8/deficiency , Matrix Metalloproteinase 8/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Paracrine Communication , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction , Stem Cells/pathology , Vascular Remodeling
4.
Breast Cancer Res ; 19(1): 33, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28330493

ABSTRACT

BACKGROUND: Normal myoepithelial cells (MECs) play an important tumour-suppressor role in the breast but display an altered phenotype in ductal carcinoma in situ (DCIS), gaining tumour-promoter functions. Matrix metalloproteinase-8 (MMP-8) is expressed by normal MECs but is lost in DCIS. This study investigated the function of MMP-8 in MECs and the impact of its loss in DCIS. METHODS: Primary normal and DCIS-associated MECs, and normal (N-1089) and DCIS-modified myoepithelial (ß6-1089) cell lines, were used to assess MMP-8 expression and function. ß6-1089 lacking MMP-8 were transfected with MMP-8 WT and catalytically inactive MMP-8 EA, and MMP-8 in N-1089 MEC was knocked down with siRNA. The effect on adhesion and migration to extracellular matrix (ECM), localisation of α6ß4 integrin to hemidesmosomes (HD), TGF-ß signalling and gelatinase activity was measured. The effect of altering MEC MMP-8 expression on tumour cell invasion was investigated in 2D and 3D organotypic models. RESULTS: Assessment of primary cells and MEC lines confirmed expression of MMP-8 in normal MEC and its loss in DCIS-MEC. Over-expression of MMP-8 WT but not MMP-8 EA in ß6-1089 cells increased adhesion to ECM proteins and reduced migration. Conversely, knock-down of MMP-8 in N-1089 reduced adhesion and increased migration. Expression of MMP-8 WT in ß6-1089 led to greater localisation of α6ß4 to HD and reduced retraction fibre formation, this being reversed by MMP-8 knock-down in N-1089. Over-expression of MMP-8 WT reduced TGF-ß signalling and gelatinolytic activity. MMP-8 knock-down enhanced TGF-ß signalling and gelatinolytic activity, which was reversed by blocking MMP-9 by knock-down or an inhibitor. MMP-8 WT but not MMP-8 EA over-expression in ß6-1089 reduced breast cancer cell invasion in 2D and 3D invasion assays, while MMP-8 knock-down in N-1089 enhanced cancer cell invasion. Staining of breast cancer cases for MMP-8 revealed a statistically significant loss of MMP-8 expression in DCIS with invasion versus pure DCIS (p = 0.001). CONCLUSIONS: These data indicate MMP-8 is a vital component of the myoepithelial tumour-suppressor function. It restores MEC interaction with the matrix, opposes TGF-ß signalling and MMP-9 proteolysis, which contributes to inhibition of tumour cell invasion. Assessment of MMP-8 expression may help to determine risk of DCIS progression.


Subject(s)
Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Matrix Metalloproteinase 8/deficiency , Biomarkers, Tumor , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Adhesion , Cell Line, Transformed , Cell Line, Tumor , Cell Movement , Cell Survival , Female , Gene Expression , Gene Knockdown Techniques , Humans , Immunohistochemistry , Integrin alpha6beta4/metabolism , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase 8/metabolism , Matrix Metalloproteinase 9/metabolism , Paracrine Communication , Protein Transport , Proteolysis , Signal Transduction , Transforming Growth Factor beta/metabolism
5.
J Cell Physiol ; 231(11): 2506-16, 2016 11.
Article in English | MEDLINE | ID: mdl-26923552

ABSTRACT

Our previous studies have shown that Dexamethasone (Dex) reduced the expression of matrix-metalloproteinases (MMPs -1,-3,-9,-13), IL-1ß and IL-6, while it significantly increased MMP-8 mRNA transcripts in a concomitant dry eye and corneal alkali burn murine model (CM). To investigate if MMP-8 induction is responsible for some of the protective effects of Dex in CM, MMP-8 knock out mice (MMP-8KO) were subjected to the CM for 2 or 5 days and topically treated either with 2 µl of 0.1% Dexamethasone (Dex), or saline QID. A separate group of C57BL/6 mice were topically treated with Dex or BSS and received either 100 nM CAM12 (MMP-8 inhibitor) or vehicle IP, QD. Here we demonstrate that topical Dex treated MMP-8KO mice subjected to CM showed reduced corneal clarity, increased expression of inflammatory mediators (IL-6, CXCL1, and MMP-1 mRNA) and increased neutrophil infiltration at 2D and 5D compared to Dex treated WT mice. C57BL/6 mice topically treated with Dex and CAM12 IP recapitulated findings seen with MMP-8KO mice. These results suggest that some of the anti-inflammatory effects of Dex are mediated through increased MMP-8 expression. J. Cell. Physiol. 231: 2506-2516, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Burns, Chemical/drug therapy , Burns, Chemical/enzymology , Cornea/pathology , Dexamethasone/therapeutic use , Dry Eye Syndromes/complications , Eye Burns/drug therapy , Eye Burns/enzymology , Matrix Metalloproteinase 8/metabolism , Alkalies , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Burns, Chemical/complications , Cornea/enzymology , Desiccation , Dexamethasone/pharmacology , Disease Models, Animal , Dry Eye Syndromes/enzymology , Eye Burns/complications , Female , Matrix Metalloproteinase 8/deficiency , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase Inhibitors/pharmacology , Matrix Metalloproteinase Inhibitors/therapeutic use , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stress, Physiological
6.
FASEB J ; 29(4): 1435-45, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25550459

ABSTRACT

Various cell types in atherosclerotic lesions express matrix metalloproteinase (MMP)-8. We investigated whether MMP-8 affects the structure and antiatherogenic function of apolipoprotein (apo) A-I, the main protein component of HDL particles. Furthermore, we studied serum lipid profiles and cholesterol efflux capacity in MMP-8-deficient mouse model. Incubation of apoA-I (28 kDa) with activated MMP-8 yielded 22 kDa and 25 kDa apoA-I fragments. Mass spectrometric analyses revealed that apoA-I was cleaved at its carboxyl-terminal part. Treatment of apoA-I and HDL with MMP-8 resulted in significant reduction (up to 84%, P < 0.001) in their ability to facilitate cholesterol efflux from cholesterol-loaded THP-1 macrophages. The cleavage of apoA-I by MMP-8 and the reduction in its cholesterol efflux capacity was inhibited by doxycycline. MMP-8-deficient mice had significantly lower serum triglyceride (TG) levels (P = 0.003) and larger HDL particles compared with wild-type (WT) mice. However, no differences were observed in the apoA-I levels or serum cholesterol efflux capacities between the mouse groups. Proteolytic modification of apoA-I by MMP-8 may impair the first steps of reverse cholesterol transport, leading to increased accumulation of cholesterol in the vessel walls. Eventually, inhibition of MMPs by doxycycline may reduce the risk for atherosclerotic vascular diseases.


Subject(s)
Apolipoprotein A-I/metabolism , Cholesterol/metabolism , Matrix Metalloproteinase 8/metabolism , Amino Acid Sequence , Animals , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/genetics , Atherosclerosis/etiology , Atherosclerosis/metabolism , Binding Sites/genetics , Biological Transport, Active , Cell Line , Cholesterol/blood , Female , Humans , Lipoproteins, HDL/metabolism , Macrophages/metabolism , Male , Matrix Metalloproteinase 8/deficiency , Matrix Metalloproteinase 8/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Proteolysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Triglycerides/blood
7.
Hepatology ; 60(4): 1409-17, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24700614

ABSTRACT

UNLABELLED: Remote ischemic preconditioning (RIPC), the repetitive transient mechanical obstruction of vessels at a limb remote to the operative site, is a novel strategy to mitigate distant organ injury associated with surgery. In the clinic, RIPC has demonstrated efficacy in protecting various organs against ischemia reperfusion (IR), but a common mechanism underlying the systemic protection has not been identified. Here, we reasoned that protection may rely on adaptive physiological responses toward local stress, as is incurred through RIPC. Standardized mouse models of partial hepatic IR and of RIPC to the femoral vascular bundle were applied. The roles of platelets, peripheral serotonin, and circulating vascular endothelial growth factor (Vegf) were studied in thrombocytopenic mice, Tph1(-) (/) (-) mice, and through neutralizing antibodies, respectively. Models of interleukin-10 (Il10) and matrix metalloproteinase 8 (Mmp8) deficiency were used to assess downstream effectors of organ protection. The protection against hepatic IR through RIPC was dependent on platelet-derived serotonin. Downstream of serotonin, systemic protection was spread through up-regulation of circulating Vegf. Both RIPC and serotonin-Vegf induced differential gene expression in target organs, with Il10 and Mmp8 displaying consistent up-regulation across all organs investigated. Concerted inhibition of both molecules abolished the protective effects of RIPC. RIPC was able to mitigate pancreatitis, indicating that it can protect beyond ischemic insults. CONCLUSIONS: We have identified a platelet-serotonin-Vegf-Il10/Mmp8 axis that mediates the protective effects of RIPC. The systemic action, the conservation of RIPC effects among mice and humans, and the protection beyond ischemic insults suggest that the platelet-dependent axis has evolved as a preemptive response to local stress, priming the body against impending harm.


Subject(s)
Blood Platelets/physiology , Ischemic Preconditioning/methods , Liver/blood supply , Reperfusion Injury/prevention & control , Reperfusion Injury/physiopathology , Signal Transduction/physiology , Animals , Disease Models, Animal , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-10/metabolism , Matrix Metalloproteinase 8/deficiency , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase 8/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Serotonin/deficiency , Serotonin/genetics , Serotonin/metabolism , Thrombocytopenia/metabolism , Thrombocytopenia/pathology , Thrombocytopenia/physiopathology , Tryptophan Hydroxylase/deficiency , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
8.
Arterioscler Thromb Vasc Biol ; 34(1): 90-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24158518

ABSTRACT

OBJECTIVE: We investigated the role of matrix metalloproteinase-8 (MMP8) in neointima formation and in vascular smooth muscle cell (VSMC) migration and proliferation. APPROACH AND RESULTS: After carotid artery wire injuring, MMP8(-/-)/apoE(-/-) mice had fewer proliferating cells in neointimal lesions and smaller lesion sizes. Ex vivo assays comparing VSMCs isolated from MMP8 knockout and wild-type mice showed that MMP8 knockout decreased proliferation and migration. Proteomics analysis revealed that a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) had lower concentrations in MMP8 knockout VSMC culture media than in MMP8 wild-type VSMC culture media. Western blot, flow cytometric, and immunocytochemical analyses showed that MMP8 knockout VSMCs contained more pro-ADAM10 but less mature ADAM10, more N-cadherin, and ß-catenin in the plasma membrane but less ß-catenin in the nucleus and less cyclin D1. Treatment of MMP8 wild-type VSMCs with an ADAM10 inhibitor, GI254023X, or siRNA knockdown of ADAM10 in MMP8 wild-type VSMCs inhibited proliferation and migration, increased N-cadherin and ß-catenin in the plasma membrane, reduced ß-catenin in the nucleus, and decreased cyclin D1 expression. Incubation of MMP8 knockout VSMCs with a recombinant ADAM10 rescued the proliferative and migratory ability of MMP8 knockout VSMCs and increased cyclin D1 expression. Furthermore, immunohistochemical analyses showed colocalization of ADAM10 with VSMCs and N-cadherin, and nuclear accumulation of ß-catenin in the neointima in apoE(-/-)/MMP8(+/+) mice. CONCLUSIONS: MMP8 enhances VSMC proliferation via an ADAM10, N-cadherin, and ß-catenin-mediated pathway and plays an important role in neointima formation.


Subject(s)
Carotid Artery Injuries/enzymology , Carotid Artery Injuries/pathology , Cell Proliferation , Matrix Metalloproteinase 8/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Neointima , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM10 Protein , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Cadherins/metabolism , Carotid Artery Injuries/genetics , Cell Movement , Cell Proliferation/drug effects , Cells, Cultured , Culture Media, Conditioned/metabolism , Cyclin D1/metabolism , Disease Models, Animal , Matrix Metalloproteinase 8/deficiency , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase Inhibitors/pharmacology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Proteomics/methods , RNA Interference , Time Factors , Transfection , Wnt Signaling Pathway , Wnt1 Protein/metabolism , beta Catenin/metabolism
9.
J Immunol ; 190(8): 4283-96, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23487425

ABSTRACT

Matrix metalloproteinase-8 (MMP-8) is a potent interstitial collagenase thought to be expressed mainly by polymorphonuclear neutrophils. To determine whether MMP-8 regulates lung inflammatory or fibrotic responses to bleomycin, we delivered bleomycin by the intratracheal route to wild-type (WT) versus Mmp-8(-/-) mice and quantified MMP-8 expression, and inflammation and fibrosis in the lung samples. Mmp-8 steady state mRNA and protein levels increase in whole lung and bronchoalveolar lavage samples when WT mice are treated with bleomycin. Activated murine lung fibroblasts express Mmp-8 in vitro. MMP-8 expression is increased in leukocytes in the lungs of patients with idiopathic pulmonary fibrosis compared with control lung samples. Compared with bleomycin-treated WT mice, bleomycin-treated Mmp-8(-/-) mice have greater lung inflammation, but reduced lung fibrosis. Whereas bleomycin-treated Mmp-8(-/-) and WT mice have similar lung levels of several pro- and antifibrotic mediators (TGF-ß, IL-13, JE, and IFN-γ), Mmp-8(-/-) mice have higher lung levels of IFN-γ-inducible protein-10 (IP-10) and MIP-1α. Genetically deleting either Ip-10 or Mip-1α in Mmp-8(-/-) mice abrogates their lung inflammatory response to bleomycin, but reconstitutes their lung fibrotic response to bleomycin. Studies of bleomycin-treated Mmp-8 bone marrow chimeric mice show that both leukocytes and lung parenchymal cells are sources of profibrotic MMP-8 during bleomycin-mediated lung fibrosis. Thus, during bleomycin-mediated lung injury, MMP-8 dampens the lung acute inflammatory response, but promotes lung fibrosis by reducing lung levels of IP-10 and MIP-1α. These data indicate therapeutic strategies to reduce lung levels of MMP-8 may limit fibroproliferative responses to injury in the human lung.


Subject(s)
Acute Lung Injury/enzymology , Bleomycin/administration & dosage , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/pathology , Matrix Metalloproteinase 8/physiology , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Animals , Disease Models, Animal , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Matrix Metalloproteinase 8/deficiency , Matrix Metalloproteinase 8/metabolism , Matrix Metalloproteinase Inhibitors/administration & dosage , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Primary Cell Culture
10.
Cardiovasc Res ; 99(1): 146-55, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23512982

ABSTRACT

AIMS: Growing evidence suggests a close association of plaque angiogenesis with atherosclerotic plaque formation and progression, and an important role of matrix metalloproteinase (MMP) in angiogenesis and atherosclerosis. We attempted to investigate the functional involvements of MMP8 in angiogenesis. METHODS AND RESULTS: Knockdown of MMP8 in human umbilical vein endothelial cells (HuVECs) with MMP-8 shRNA lentivirus resulted in a decrease in in vitro capillary-like network formation, cell proliferation and migration, and impaired its capacity of in vivo angiogenesis. Less nuclear accumulation of ß-catenin and lower ß-catenin target gene expression levels was observed in the HuVECs expressing lower levels of endogenous MMP8. Knockdown of endogenous MMP8 in HuVECs down-regulated platelet/endothelial cell adhesion molecule-1 (PECAM-1) expression by converting less angiotensin I to II, which is an inducer for PECAM-1 gene expression. Aortic rings isolated from MMP8(-/-)/apoE(-/-) mice had less endothelial cell sprouting, and endothelial cells in MMP8(-/-)/apoE(-/-) mice had a lower ability to migrate into Matrigel plugs and less capacity of proliferation and angiogenesis. Moreover, immunohistochemical analyses revealed that MMP8 was expressed in microvessels within human atherosclerotic plaques and aneurysm. Finally, analyses of MMP8(-/-)/apoE(-/-) and MMP8(+/+)/apoE(-/-) mice fed a Western diet for 12 weeks showed that MMP8-deficient mice had small lesion size and less endothelial cells within atherosclerotic lesions. CONCLUSION: We demonstrated for the first time that MMP8 plays an important role in angiogenesis in vitro and in vivo. Our findings provide new insights into the molecular mechanisms of plaque angiogenesis and suggest that MMP8 is a potential therapeutic target of cardiovascular diseases.


Subject(s)
Human Umbilical Vein Endothelial Cells/enzymology , Matrix Metalloproteinase 8/metabolism , Neovascularization, Physiologic , Aneurysm/enzymology , Aneurysm/pathology , Angiotensin I/metabolism , Angiotensin II/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/enzymology , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Gene Knockdown Techniques , Humans , Matrix Metalloproteinase 8/deficiency , Matrix Metalloproteinase 8/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic , Plaque, Atherosclerotic , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA Interference , Time Factors , Transfection , beta Catenin/metabolism
11.
Circ Res ; 112(1): 35-47, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23071158

ABSTRACT

RATIONALE: Accumulating evidence indicates that stem/progenitor cells (SPCs) represent an important source of cells in atheromas and contribute to lesion formation and progression. OBJECTIVE: We investigated whether matrix metalloproteinase-8 (MMP8) played a role in SPC migration and their recruitment into atheromas. METHODS AND RESULTS: We found that SPCs in atheromas expressed MMP8 and that MMP8 knockout significantly reduced SPC numbers in atherosclerotic lesions in apolipoprotein E (ApoE)-deficient mice fed a Western diet. Further in vivo experiments showed that ApoE(-/-)/MMP8(-/-) mice injected with stem cells isolated from bone marrows of ApoE(-/-)/MMP8(-/-) mice had fewer SPCs in atheromas and smaller lesions than ApoE(-/-)/MMP8(-/-) mice injected with stem cells isolated from bone marrows of ApoE(-/-)/MMP8(+/+) mice. Ex vivo experiments showed that MMP8 deficiency inhibited the ability of SPCs to migrate from the arterial lumen and the adventitia into atherosclerotic lesions. In vitro assays indicated that MMP8 facilitated SPC migration across endothelial cells and through Matrigel or collagen I. We also found that MMP8 cleaved a-disintegrin-and-metalloproteinase-domain-10 and that MMP8 deficiency reduced mature a-disintegrin-and-metalloproteinase-domain-10 on SPCs. Knockdown of MMP8 or incubation with the a-disintegrin-and-metalloproteinase-domain-10 inhibitor GI254023X decreased E-cadherin shedding on SPCs. The decrease in migratory ability of SPCs with MMP8 knockdown was reduced by incubation of such cells with culture supernatant from SPCs without MMP8 knockdown, and this compensatory effect was abolished by an antibody against soluble E-cadherin. CONCLUSIONS: MMP8 plays an important role in SPC migration and their recruitment into atherosclerotic lesions.


Subject(s)
Aorta/enzymology , Aortic Diseases/enzymology , Atherosclerosis/enzymology , Cell Movement , Matrix Metalloproteinase 8/metabolism , Stem Cells/enzymology , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/metabolism , Animals , Antigens, Ly/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Biomarkers/metabolism , Cadherins/metabolism , Cells, Cultured , Collagen Type I/metabolism , Dipeptides/pharmacology , Disease Models, Animal , Endothelial Cells/enzymology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hydroxamic Acids/pharmacology , Male , Matrix Metalloproteinase 8/deficiency , Matrix Metalloproteinase 8/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Plaque, Atherosclerotic , Protease Inhibitors/pharmacology , RNA Interference , Stem Cell Transplantation , Stem Cells/drug effects , Stem Cells/pathology , Time Factors , Transfection
12.
PLoS One ; 7(6): e39940, 2012.
Article in English | MEDLINE | ID: mdl-22768176

ABSTRACT

Matrix metalloproteinase-8, released mainly from neutrophils, is a critical regulator of the inflammatory response by its ability to cleave multiple mediators. Herein, we report the results of a model of endotoxemia after intraperitoneal LPS injection in mice lacking MMP-8 and their wildtype counterparts. Control, saline-treated animals showed no differences between genotypes. However, there was an increased lung inflammatory response, with a prominent neutrophilic infiltration in mutant animals after LPS treatment. Using a proteomic approach, we identify alarmins S100A8 and S100A9 as two of the main differences between genotypes. Mice lacking MMP-8 showed a significant increase in these two molecules in lung homogenates, but not in spleen and serum. Mice lacking MMP-8 also showed an increase in MIP-1α levels and a marked activation of the non-canonical NF-κB pathway, with no differences in CXC-chemokines such as MIP-2 or LIX. These results show that MMP-8 can modulate the levels of S100A8 and S100A9 and its absence promotes the lung inflammatory response during endotoxemia.


Subject(s)
Calgranulin A/metabolism , Calgranulin B/metabolism , Endotoxemia/enzymology , Matrix Metalloproteinase 8/deficiency , Pneumonia/enzymology , Receptors, Immunologic/metabolism , Toll-Like Receptors/metabolism , Animals , Endotoxemia/complications , Endotoxemia/immunology , Endotoxemia/pathology , Genotype , Ligands , Lipopolysaccharides/pharmacology , Lung/drug effects , Lung/microbiology , Lung/pathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 8/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Pneumonia/complications , Pneumonia/immunology , Pneumonia/pathology , Proteomics , Receptor for Advanced Glycation End Products , Signal Transduction/drug effects
13.
J Periodontal Res ; 46(1): 58-66, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20731768

ABSTRACT

BACKGROUND AND OBJECTIVE: Matrix metalloproteinase-8 (MMP-8) is a central mediator in chronic periodontitis. Recently developed MMP-8-deficient mice show an impaired polymorphonuclear neutrophil response and more severe alveolar bone loss in Porphyromonas gingivalis-induced experimental periodontitis. The main mediators involved in neutrophil and monocyte/macrophage recruitment and in bone loss include lipopolysaccharide-induced CXC chemokine (LIX/CXCL5), stromal-derived factor-1/CXC chemokine ligand 12 (SDF1/CXCL12) and RANKL. Therefore, the aim of this study was to characterize the expression of LIX/CXCL5, SDF1/CXCL12 and RANKL in Porphyromonas gingivalis-induced experimental periodontitis in MMP-8⁻/⁻ (knockout) and wild-type mice. MATERIAL AND METHODS: MMP-8 null and WT P. gingivalis-infected and uninfected mice were included. Histopathological changes were assessed and LIX/CXCL5, SDF1/CXCL12 and RANKL were immunodetected and quantified. RESULTS: Typical histopathological features of chronic periodontitis were seen in P. gingivalis-infected groups. LIX/CXCL5 expression was restricted to the gingival papilla in all four groups. Significantly lower expression of LIX/CXCL5 was seen in the knockout group compared with the wild-type infected group (p < 0.05). SDF1/CXCL12 and RANKL expression was mainly localized to the alveolar crest, including inflammatory leukocytes, vascular endothelium, osteoblasts and osteoclasts. Significant increases of SDF1/CXCL12 and RANKL were seen in both knockout and wild-type P. gingivalis-infected groups compared with uninfected groups (p < 0.05). CONCLUSION: RANKL and SDF1/CXCL12 are up-regulated in P. gingivalis-induced periodontitis and they appear to be associated with the pathogenesis of the disease. MMP-8 is associated with a reduced expression of LIX/CXCL5 in the P. gingivalis-induced experimental periodontitis model.


Subject(s)
Alveolar Bone Loss/metabolism , Chemokine CXCL5/biosynthesis , Chronic Periodontitis/metabolism , Matrix Metalloproteinase 8/metabolism , Alveolar Bone Loss/microbiology , Animals , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/genetics , Chemokine CXCL5/genetics , Chronic Periodontitis/microbiology , Gene Expression Regulation , Lipopolysaccharides/pharmacology , Male , Matrix Metalloproteinase 8/deficiency , Matrix Metalloproteinase 8/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Porphyromonas gingivalis , RANK Ligand/biosynthesis , RANK Ligand/genetics
14.
Arthritis Res Ther ; 12(6): R224, 2010.
Article in English | MEDLINE | ID: mdl-21190566

ABSTRACT

INTRODUCTION: Rheumatoid arthritis is an autoimmune disease in which joint inflammation leads to progressive cartilage and bone erosion. Matrix metalloproteinases (MMPs) implicated in homeostasis of the extracellular matrix play a central role in cartilage degradation. However, the role of specific MMPs in arthritis pathogenesis is largely unknown. The aim of the present study was to investigate the role of Mmp-8 (collagenase-2) in an arthritis model. METHODS: Arthritis was induced in Mmp8-deficient and wildtype mice by K/BxN serum transfer. Arthritis severity was measured by a clinical index and ankle sections were scored for synovial inflammation, cartilage damage and bone erosion. cDNA microarray analysis, real-time PCR and western blot were performed to identify differential changes in gene expression between mice lacking Mmp8 and controls. RESULTS: Mmp8 deficiency increased the severity of arthritis, although the incidence of disease was similar in control and deficient mice. Increased clinical score was associated with exacerbated synovial inflammation and bone erosion. We also found that the absence of Mmp8 led to increased expression of IL-1ß, pentraxin-3 (PTX3) and prokineticin receptor 2 (PROKR2) in arthritic mice joints. CONCLUSIONS: Lack of Mmp-8 is accompanied by exacerbated synovial inflammation and bone erosion in the K/BxN serum-transfer arthritis model, indicating that this Mmp has a protective role in arthritis.


Subject(s)
Arthritis, Experimental/enzymology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/pathology , Matrix Metalloproteinase 8/deficiency , Animals , Ankle Joint/pathology , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Blotting, Western , Bone and Bones/metabolism , Bone and Bones/pathology , Disease Models, Animal , Inflammation/enzymology , Inflammation/pathology , Matrix Metalloproteinase 8/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
15.
Arthritis Rheum ; 62(12): 3645-55, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21120997

ABSTRACT

OBJECTIVE: Neutrophil accumulation is balanced by both cell infiltration and cell clearance, the controls of which are pivotal in the pathogenesis of rheumatoid arthritis (RA) and other chronic inflammatory diseases. Of the neutrophil-specific proteases, matrix metalloproteinase 8 (MMP-8; also known as neutrophil collagenase or collagenase 2) is traditionally viewed as being crucial for collagen degradation and hence cell migration and infiltration. This study was undertaken to examine the role of MMP-8 in a murine model of spontaneous RA. METHODS: MMP-8(-/-) mice were backcrossed onto the Fas-defective MRL/lpr background, a mouse strain characterized by systemic autoimmunity including spontaneous autoimmune arthritis. Arthritis was induced with Freund's complete adjuvant and clinical disease and histologic parameters were assessed. RESULTS: MMP-8(-/-) mice had earlier and more severe joint inflammation than their MMP-8(+/+) counterparts, coupled with a massive accumulation of neutrophils in synovial tissue, an unexpected result considering the commonly held view that MMP-8 has important extracellular matrix-degradative functions. Protease and protease inhibitor analysis of MMP-8(-/-) mouse neutrophils by CLIP-CHIP microarray revealed very little additional change in protease levels except for low expression of the apoptosis initiator caspase 11. This was confirmed at the protein level in unstimulated, lipopolysaccharide-treated, and interferon-γ-treated MMP-8(-/-) mouse neutrophils. Downstream of caspase 11, the activity of the apoptosis executioner caspase 3 was consequently reduced in MMP-8(-/-) mouse neutrophils, translating to reduced neutrophil apoptosis and cell accumulation compared with wild-type mouse cells. CONCLUSION: Our findings indicate that MMP-8 is not essential for neutrophil migration in arthritis and likely other autoimmune diseases. Rather, MMP-8 is important for normal rates of neutrophil apoptosis and hence regulates cell clearance. Because MMP-8 deficiency leads to an exaggerated accumulation of neutrophil infiltrates due to delayed apoptosis and concurrent pathologic changes associated with dramatically increased neutrophil infiltration, MMP-8 is antiinflammatory and therefore a drug antitarget in the treatment of arthritis.


Subject(s)
Apoptosis/physiology , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Caspases/metabolism , Matrix Metalloproteinase 8/deficiency , Neutrophils/pathology , Animals , Apoptosis/drug effects , Caspases, Initiator , Cell Movement/drug effects , Disease Models, Animal , Female , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Male , Matrix Metalloproteinase 8/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/drug effects , Neutrophils/metabolism , Synovial Membrane/metabolism , Synovial Membrane/pathology
16.
J Immunol ; 184(3): 1575-88, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20042585

ABSTRACT

To determine the role of matrix metalloproteinase-8 (MMP-8) in acute lung injury (ALI), we delivered LPS or bleomycin by the intratracheal route to MMP-8(-/-) mice versus wild-type (WT) mice or subjected the mice to hyperoxia (95% O(2)) and measured lung inflammation and injury at intervals. MMP-8(-/-) mice with ALI had greater increases in lung polymorphonuclear neutrophils (PMNs) and macrophage counts, measures of alveolar capillary barrier injury, lung elastance, and mortality than WT mice with ALI. Bronchoalveolar lavage fluid (BALF) from LPS-treated MMP-8(-/-) mice had more MIP-1alpha than BALF from LPS-treated WT mice, but similar levels of other pro- and anti-inflammatory mediators. MIP-1alpha(-/-) mice with ALI had less acute lung inflammation and injury than WT mice with ALI, confirming that MIP-1alpha promotes acute lung inflammation and injury in mice. Genetically deleting MIP-1alpha in MMP-8(-/-) mice reduced the increased lung inflammation and injury and mortality in MMP-8(-/-) mice with ALI. Soluble MMP-8 cleaved and inactivated MIP-1alpha in vitro, but membrane-bound MMP-8 on activated PMNs had greater MIP-1alpha-degrading activity than soluble MMP-8. High levels of membrane-bound MMP-8 were detected on lung PMNs from LPS-treated WT mice, but soluble, active MMP-8 was not detected in BALF samples. Thus, MMP-8 has novel roles in restraining lung inflammation and in limiting alveolar capillary barrier injury during ALI in mice by inactivating MIP-1alpha. In addition, membrane-bound MMP-8 on activated lung PMNs is likely to be the key bioactive form of the enzyme that limits lung inflammation and alveolar capillary barrier injury during ALI.


Subject(s)
Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Chemokine CCL3/antagonists & inhibitors , Chemokine CCL3/metabolism , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Matrix Metalloproteinase 8/physiology , Acute Lung Injury/enzymology , Animals , Bleomycin/toxicity , Chemokine CCL3/deficiency , Enzyme Activation/immunology , Humans , Lung/enzymology , Lung/pathology , Matrix Metalloproteinase 8/deficiency , Membrane Proteins/deficiency , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Activation/immunology , Neutrophils/enzymology , Neutrophils/pathology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity/immunology
17.
J Leukoc Biol ; 87(1): 69-77, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19801498

ABSTRACT

Increased levels of MMP-8 (neutrophil collagenase) have been reported in OB, but the biological role of MMP-8 in OB is not known. MMP-8 is an interstitial collagenase highly expressed by polymorphonuclear leukocytes, which are prominent in early OB. Here, we show that MMP-8 promotes migration of PMNs through the collagen-rich matrix in a mouse heterotopic airway transplant model of OB. Overall, MMP-8(-/-) mice had significantly fewer PMNs in the airway lumen 2 and 14 days post-transplantation, and the percentage of PMNs traversing the matrix to the lumen was decreased markedly in the MMP-8(-/-) compared with WT mice at 14 days. There were significantly more PMNs outside of the lumen in the ECM in the MMP-8(-/-) mice compared with WT mice. In vitro, significantly fewer MMP-8(-/-) PMNs migrated through 3D cross-linked collagen gels than WT PMNs. MMP inhibitor GM6001 was also able to impede migration of WT PMNs through collagen gels. The decreased migration was likely a result of pericollagenase activity of MMP-8, as WT PMNs expressing MMP-8 were not able to migrate effectively through collagen that was resistant to the collagenase. Protection from OB was seen in the MMP-8(-/-) mice, as the airway lumen had significantly less obliteration and collagen deposition, suggesting that MMP-8 plays an important role in the pathogenesis of OB.


Subject(s)
Bronchiolitis Obliterans/immunology , Chemotaxis, Leukocyte/physiology , Matrix Metalloproteinase 8/physiology , Animals , Blood Cells/physiology , Bone Marrow Cells/physiology , Bronchiolitis Obliterans/enzymology , Cells, Cultured/physiology , Collagen , Extracellular Matrix , Female , Gels , Matrix Metalloproteinase 8/deficiency , Matrix Metalloproteinase 8/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/physiology , Trachea/transplantation , Transplantation, Heterotopic
18.
Am J Respir Cell Mol Biol ; 43(5): 555-63, 2010 Nov.
Article in English | MEDLINE | ID: mdl-19995943

ABSTRACT

Mechanical ventilation is a life-saving therapy that can also damage the lungs. Ventilator-induced lung injury (VILI) promotes inflammation and up-regulates matrix metalloproteinases (MMPs). Among these enzymes, MMP-8 is involved in the onset of inflammation by processing different immune mediators. To clarify the role of MMP-8 in a model of VILI and their relevance as a therapeutic target, we ventilated wild-type and MMP-8-deficient mice with low or high pressures for 2 hours. There were no significant differences after low-pressure ventilation between wild-type and knockout animals. However, lack of MMP-8 results in better gas exchange, decreased lung edema and permeability, and diminished histological injury after high-pressure ventilation. Mmp8(-/-) mice had a different immune response to injurious ventilation, with decreased neutrophilic infiltration, lower levels of IFN-γ and chemokines (LPS-induced CXC chemokine, macrophage inflammatory protein-2), and significant increases in anti-inflammatory cytokines (IL-4, IL-10) in lung tissue and bronchoalveolar lavage fluid. There were no differences in MMP-2, MMP-9, or tissue inhibitor of metalloproteinase-1 between wild-type and knockout mice. These results were confirmed by showing a similar protective effect in wild-type mice treated with a selective MMP-8 inhibitor. We conclude that MMP-8 promotes acute inflammation after ventilation with high pressures, and its short-term inhibition could be a therapeutic goal to limit VILI.


Subject(s)
Matrix Metalloproteinase 8/deficiency , Matrix Metalloproteinase Inhibitors , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/prevention & control , Animals , Biomarkers/metabolism , Cytokines/pharmacology , Enzyme Inhibitors/pharmacology , Hydrogen-Ion Concentration/drug effects , Lung/enzymology , Lung/immunology , Lung/pathology , Lung/physiopathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 8/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Pulmonary Gas Exchange/drug effects , Tissue Inhibitor of Metalloproteinase-1/metabolism , Ventilator-Induced Lung Injury/immunology , Ventilator-Induced Lung Injury/physiopathology
20.
Circ Res ; 105(9): 921-9, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19745165

ABSTRACT

RATIONALE: Atherosclerotic lesions express matrix metalloproteinase (MMP)8, which possesses proteolytic activity on matrix proteins particularly fibrillar collagens and on nonmatrix proteins such as angiotensin (Ang) I. OBJECTIVE: We studied whether MMP8 plays a role in atherogenesis. METHODS AND RESULTS: In atherosclerosis-prone apolipoprotein E-deficient mice, inactivating MMP8 resulted in a substantial reduction in atherosclerotic lesion formation. Immunohistochemical examinations showed that atherosclerotic lesions in MMP8-deficient mice had significantly fewer macrophages but increased collagen content. In line with results of in vitro assays showing that Ang I cleavage by MMP8 generated Ang II, MMP8 knockout mice had lower Ang II levels and lower blood pressure. In addition, we found that products of Ang I cleavage by MMP8 increased vascular cell adhesion molecule (VCAM)-1 expression and that MMP8-deficient mice had reduced VCAM-1 expression in atherosclerotic lesions. Intravital microscopy analysis showed that leukocyte rolling and adhesion on vascular endothelium was reduced in MMP8 knockout mice. Furthermore, we detected an association between MMP8 gene variation and extent of coronary atherosclerosis in patients with coronary artery disease. A relationship among MMP8 gene variation, plasma VCAM-1 level, and atherosclerosis progression was also observed in a population-based, prospective study. CONCLUSIONS: These results indicate that MMP8 is an important player in atherosclerosis.


Subject(s)
Aortic Diseases/enzymology , Atherosclerosis/enzymology , Carotid Artery Diseases/enzymology , Coronary Stenosis/enzymology , Matrix Metalloproteinase 8/metabolism , Angiotensin II/metabolism , Animals , Aortic Diseases/genetics , Aortic Diseases/physiopathology , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/physiopathology , Atherosclerosis/prevention & control , Blood Pressure , Carotid Artery Diseases/genetics , Carotid Artery Diseases/prevention & control , Collagen/metabolism , Coronary Stenosis/genetics , Coronary Stenosis/prevention & control , Disease Models, Animal , Disease Progression , Endothelium, Vascular/metabolism , Gene Frequency , Genetic Predisposition to Disease , Humans , Leukocyte Rolling , Macrophages/metabolism , Male , Matrix Metalloproteinase 8/deficiency , Matrix Metalloproteinase 8/genetics , Mice , Mice, Knockout , Polymorphism, Single Nucleotide , Prospective Studies , Severity of Illness Index , Vascular Cell Adhesion Molecule-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...