Basic & Clinical Pharmacology & Toxicology, 2014, 115, 301–314

Doi: 10.1111/bcpt.12282

MiniReview

Matrix Metalloproteinases are Involved in Cardiovascular Diseases Aline Azevedo1, Alejandro F. Prado1, Raquel C. Antonio1, Joao P. Issa2 and Raquel F. Gerlach1,2 1

Department of Pharmacology, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil and 2Department of Morphology and Physiology, Faculty of Dentistry of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil (Received 15 June 2013; Accepted 26 June 2014) Abstract: This MiniReview describes the essential biochemical and molecular aspects of matrix metalloproteinases (MMPs) and briefly discusses how they engage in different diseases, with particular emphasis on cardiovascular diseases. There is compelling scientific evidence that many MMPs, especially MMP-2, play important roles in the development of cardiovascular diseases; inhibition of these enzymes is beneficial to many cardiovascular conditions, sometimes precluding or postponing end-organ damage and fatal outcomes. Conducting comprehensive discussions and further studies on how MMPs participate in cardiovascular diseases is important, because inhibition of these enzymes may be an alternative or an adjuvant for current cardiovascular disease therapy.

General Aspects of MMPs Matrix metalloproteinases also called matrixins consist of endopeptidases that participate in extracellular matrix (ECM) degradation; they represent the main group of proteases implicated in this biological process. MMPs play essential physiological roles in the organism, including embryonic development, morphogenesis, angiogenesis, ovulation, cervical dilation, bone tissue remodelling, wound healing and apoptosis [1,2]. MMPs also act in cardiovascular remodelling and have a distinct spatial and temporal role that ensures normal physiology of the heart [3] and vasculature [4]. Genetic alterations in these enzymes and MMP expression are related to the pathological processes of arthritis, fibrosis, tumour growth and metastasis, nephritis, neurological and periodontal diseases, hypertension and atherosclerosis [2]. To date, scientists have identified 24 MMP genes in human beings, most of which consist of multidomain proteins [1]. It is possible to regulate the MMP activities at different levels; gene expression regulation, zymogens activation and active enzymes inhibition by specific inhibitors underlie this regulation [5]. Members of the Matrix Metalloproteinase Family In 1962, Gross and Lapiere reported the activity of an MMP for the first time: it was the collagenolytic activity verified in tadpole tissues undergoing metamorphosis [6]. In 1975, Woolley et al. [7] purified the first human collagenase from

Author for correspondence: Racquel F. Gerlach, FORP/USP, Avenida do Cafe, S/N, 14040-904 Ribeir~ao Preto, SP, Brazil (fax +55 16 36024102, e-mail [email protected]).

rheumatoid synovium; this enzyme exhibited properties similar to those of the tadpole collagenase. Currently, MMPs comprise a family of 26 enzymes that occur in human tissues as well as organisms such as the fruit fly (Drosophila melanogaster) [8], a nematode (Caenorhabditis elegans) [9], sea urchin (Paracentrotus lividus) [10], hydra (Hydra vulgaris) [11] and plants (Arabidopsis thaliana) [12]. Matrix metalloproteinases -4, -5 and -6 do not exist, since they had been proposed, and shortly later identified as one of the MMPs that had already been described. MMP classification depends on their substrate specificity, primary structure and location in the cell; they are categorized as collagenases, gelatinases, stromelysins, matrilysins and membrane-type (MT) MMPs. Three collagenases have been identified so far: collagenase 1 (MMP-1), collagenase 2 (MMP-8) and collagenase 3 (MMP13). Gelatinases A (MMP-2) and B (MMP-9) constitute the group of gelatinases. MMP-3, MMP-10 and MMP-11 correspond to stromelysins 1, 2 and 3, respectively. MMP-7 and MMP-26 belong to the group of matrilysins, the smallest MMPs. Two types of MT-MMPs exist – four type I transmembrane proteins (MMP-14, MMP-15, MMP-16 and MMP-24) and two glycosylphosphatidylinositol (GPI)-anchored proteins (MMP-17 and MMP-25). MMP-12, MMP-19, MMP-20, MMP21, MMP-23, MMP-27 and MMP-28 have not been catalogued in any of the subgroups mentioned above. Structure and Function Matrix metalloproteinases are synthesized as zymogens; they can be secreted or membrane-bound. Subsequent MMP processing and activation allow for controlled substrate degradation.

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)

MiniReview

ALINE AZEVEDO ET AL.

302

MMPs consist of multidomain zinc metalloproteinases; they share a common domain structure composed of a signal peptide, a propeptide, a catalytic domain, a hinge region and, in the majority of the cases, a C-terminal domain [2]. The propeptide domain (about 80 amino acids) contains a conserved cysteine residue, which interacts with the zinc ion in the catalytic site and maintains the protein in its inactive state. To activate MMPs, it is necessary to open this so-called cysteine ‘switch’ [13]. The MMP catalytic domain (about 170 amino acids long) displays a zinc-binding motif HEXGHXXGXXH characteristic of the metzincin superfamily of proteinases, followed by an invariant methionine involved in a structural feature called ‘Met-turn’ [14]. Mechanism of Activation To activate MMPs, it is necessary to dissociate the interaction between the cysteine residue of the propeptide domain and the zinc ion of the catalytic domain [13]. Many agents can activate MMPs, including detergents (sodium dodecyl Sulphate, for instance), chaotropic agents, chemicals like HOCl (hypochlorous acid) and mercurial compounds and enzymes (trypsin and plasmin). Physical agents can also open up the MMP structure and expose the zinc ion. Substances that react with sulfhydryl groups (NEM – N-ethylmaleimide, GSSG – oxidized glutathione, HOCl, APMA – amino-phenyl mercuric acetate) will act in two steps. Firstly, they will interact with the cysteine of the MMP propeptide, which will result in MMP autolysis and activation. Proteolytic enzymes (trypsin, plasmin) can cleave this propetide, without affecting its cysteine residue. In a second step, the activated molecules will catalyse their autolysis, permanently removing the MMP propeptide and activating the MMP [15]. Since the beginning of the research into MMPs, many studies have taken advantage of the fact that different agents and proteinases activate MMPs. For example, gelatin zymography, which identifies gelatinase activity in gels, has been used for decades; it evidences the activity of different MMP forms (containing the catalytic domain) after SDS is washed off. Because enzyme exposure to SDS denatures the protein and dissociates the interaction between the propeptide and the catalytic domain, the zymograms reveal MMP forms that bear the propeptide domain and exhibit catalytic activity. The MMP molecules renature upon SDS removal, but the propeptide no longer interacts with the catalytic domain. Thus, even ‘inactive’ (MMPs containing the propeptide) enzymes appear on the zymograms. The common mechanism that activates latent MMP forms in tissues probably involves a cascade of proteolysis that includes other proteinases, such as other MMPs, as well as proteinases belonging to other classes. Cell-associated plasmin generation by urokinase-like plasminogen activator engages in one of the mechanisms that activate MMPs [16]. Active cell surface MTI-MMP (MMP-14) also activates MMPs. It cleaves MMP-2, potentiating further self-cleavage reactions. MMP-14 may also activate MMP-13 directly. A more likely event is that MMP-2 activates MMP-13, and then, MMP-2, MMP-3

and MMP-13 activate MMP-9. MMP-3 can also activate MMP-13. The generation of partially active or totally active MMPs allows a cascade of cleavages that generate fully active enzymes [17,18]. Matrix metalloproteinase-2 activation involves the formation of a complex of MMP-2/tissue inhibitor of metalloproteinase 2 (TIMP-2) and MMP-14 on the cell surface. For MMP-14 to catalyse proMMP-2 activation, proMMP-2 has to exist as a complex with TIMP-2, in which the MMP inhibitor binds to the proMMP-2 hemopexin domain [19–22]. In the currently accepted model of MMP-2 activation, two MMP-14 molecules join in the activator complex. The proteolytic site of one of the MMP-14 molecules binds to TIMP-2, functioning as a bridge that poises proMMP-2 for activation by a neighbouring MMP-14 molecule not yet inactivated by TIMP-2 [23]. This MMP-14/TIMP-2 complex acts as a receptor, to bind proMMP-2 to the cell surface through interaction of the exposed TIMP-2 C-terminal domain with the proMMP-2 PEX domain. MMP-14 forms a homophilic complex on the cell surface via the PEX domain, which facilitates proMMP-2 activation [24]. Autolytic cleavage by MMP-2 completes the activation, to give a lower molecular form of 64 kDa, without the propeptide domain [25]. The proteolytic removal of the propeptide region disturbs the interaction of cysteine with zinc in the propeptide and exposes the catalytic domain. It has been shown that peroxynitrite (ONOO-) activates proMMP-2 without removing the propeptide, to maintain the same molecular weight of inactive MMP-2 (72 kDa). It was also proposed that ONOO and cellular glutathione (GSH) react with the cysteine residue in the conserved domain; Sglutathiolation takes place, disrupting the thiolate bond with the catalytic Zn2+ ion and furnishing an active 72-kDa (fulllength) MMP-2 [26]. Glutathiolation also activates MMP-1, MMP-8 and MMP-9 [27]. Other Structural Considerations A flexible proline-rich hinge region follows the catalytic domain in all the MMP family members, except for MMP-7 (matrilysin-1), MMP-23 (CA-MMP) and MMP-26 (matrilysin2). Thereafter (towards the C-terminal end), most MMPs present another large domain known as the hemopexin-binding domain (about 210 amino acids), which probably operates during substrate recognition and MMP activity control. This is the case of collagenases MMP-1, MMP-8, MMP-13 and MMP-18, where the hemopexin domain is required for the triple helical interstitial collagen cleavage [28]. The hemopexin domain is also important for interactions between two MMP14 molecules during the pro-MMP-2 cell surface activation [24]. The gelatinases MMP-2 and MMP-9 have an insert of three fibronectin type II repeats in the catalytic domain. This insert generates a collagen-binding domain that binds and degrades type IV collagen or denatured collagens (gelatins) [29,30], which accounts for the high affinity of these MMPs for gelatin. This feature underlies the MMP-2 and MMP-9 purification process – an affinity column that contains gelatin bound to

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)

MiniReview

MATRIX METALLOPROTEINASES ARE INVOLVED IN CARDIOVASCULAR DISEASES

sepharose beads is the first choice to obtain pure MMP-2 and MMP-9. Recently, our group has successfully expressed and purified the full-length human recombinant of MMP-2 from bacteria, to achieve a highly soluble and functional protein for in vitro and in vivo studies [31]. Individual MMPs vary; for example, MT-MMPs either have a transmembrane domain and cytoplasmic tail at the C-terminus (MMP-14, MMP-15, MMP-16 and MMP -24) or anchor to GPI (MMP-17 and MMP-25). They have a furin recognition sequence RX[R/K]R at the propeptide C-terminus, so they undergo intracellular activation and probably express active enzymes on the cell surface [1]. Regulation of MMP Expression and Activity Gene transcription, post-translational control and interaction with endogenous inhibitors (TIMPs) regulate MMP expression and activity at different levels. At the transcriptional level, a number of MMP promoters contain cis-elements, allowing a set of trans-activators including AP-1, PEA3, Sp-1, b-catenin/ Tcf-4 and NF-kB to regulate MMP gene expression. The MMP-2/MMP-9 (gelatinases) or MMP-1/MMP-8 (collagenases) promoters have distinct compositions. The majority contains a TATA box and an AP-1-binding site as well as an upstream PEA3-binding site that is often adjacent to the AP-1binding site [32]. The MMP-8, -11 and -21 promoters also contain a TATA box, but they lack a proximal AP-1 site. On the other hand, promoters of other MMPs, including MMP-2, -14 and -28, do not harbour a TATA box. Hence, MMP transcription from them starts at multiple sites and relies mainly on the ubiquitous Sp-1 family of transcription factors, which bind to a proximal GC box. Studies have demonstrated that a functional AP-1 site mediates MMP-2 transcription in cardiac cells through binding of distinctive Fra1-JunB and FosB-JunB heterodimers [33]. This report was the first to state that defined members of the Fos and Jun transcription factor families specifically regulate this gene under conditions relevant to critical pathophysiological processes. Alfonso-Jaume et al. [34] showed that induction (via oxidant stress) of discrete AP1 transcription factor components enhances MMP-2 transcription and translation following ischaemia/reperfusion injury. As for the post-translational control, phosphorylation may modify MMP-2 activity. Indeed, human recombinant MMP-2 can undergo in vitro phosphorylation in the presence of protein kinase C (PKC), with changes in five different residues [35]. The TIMPs can also inhibit MMPs in a 1:1 stoichiometric ratio. Four types of TIMPs exist: TIMP-1, -2, -3 and -4. TIMPs are involved in cardiac fibrosis, angiogenesis and apoptosis [36]. Cardiac enriched TIMP-4 plays an important role in matrix remodeling in myocardial infarct (MI), because it may inhibit MMP-9 activity [37]. An MMP/TIMP imbalance participates in structural and functional changes in hypertensive heart disease [38]. Matrix metalloproteinases do not exist in the extracellular space only. MMP-3 occurs in the cell nucleus. Authors have also proposed that MMP activity in the nucleus of tumour

303

cells might lead to cell death [39]. MMP-2 exists in mitochondria and the endoplasmatic reticulum [40], as well as in the nucleus [41] and in the sarcomeres of cardiomyocytes [42– 45]. Little information exists about the nuclear localization sequence in MMPs or about the biological mechanism that regulates MMP entry and activation in the nucleus. One report has shown that MMP-2 present in the nucleus of cardiac myocytes engages in protein degradation during DNA repair of poly (ADP-ribose) involved in apoptosis [41]. MMP and Cardiovascular Diseases Cardiovascular diseases are the leading causes of morbidity and mortality worldwide (http://www.who.int/mediacentre/factsheets/fs317/en/index.html/March 2013). MI and sudden death due to chronic diseases such as vascular atherosclerosis, heart failure, tissue remodeling and inflammation take central roles in these pathological processes. Matrix metalloproteinase activation alters the architecture of the plaque and may directly help to disrupt plaques [46–49]. Higher MMP activity underlies aneurysms progression [50,51] and plaque rupture in coronary artery disease [52–54]. MMPs and aneurysm. Aneurysm consists in destruction and functional loss of elastin in the aortic medium. MMP plays a significant role in weakening the ECM components (elastin, collagen, fibronectin and proteoglycans), thus harming the aortic wall. MMP-1 [55], MMP-2 [55,56], MMP-3 [55], MMP-8 [57] and MMP-9 [51,56] levels are elevated in aneurysm. TIMP-3 knockout reduces collagen and elastin and raises proteolytic activities, to generate aneurysm 4 weeks after angiotensin II infusion [58]. On the other hand, TIMP-2 knockout attenuates aneurysm progression, because it plays a part in MMP-2 activation [59]. Animals transgenic for MMP-2 display increased mid-ventricular coronary luminal areas coupled with foci of aneurysmal dilation, ectasia and perivascular fibrosis [60]. MMP-12 knockout mice present attenuated aneurysm growth [61]. MMP-9 or MMP-12 protects apolipoprotein E-deficient (Apo E / ) mice against atherosclerotic plaque destruction and ectasia [62]. MMPs and diabetes. In addition, MMPs are involved in other cardiovascular diseases like diabetes. Diabetic patients exhibit impaired glucose metabolism, and the ensuing hyperglycaemia results in many complications, including vasculature damage and inability to heal wounds. In these patients, vascular damage culminates in ischaemia, which in turn contributes to the persistence of wounds. In other words, vascular damage leads to inflammation and triggers reactive oxygen species (ROS) production, which harms the ECM and prevents wound closure [63,64]. Analysis of MMP-2 and MMP-9 expression in the left ventricle (LV) of the heart of diabetic animals revealed MMP-2 down-regulation and significant MMP-9 up-regulation [65].

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)

304

ALINE AZEVEDO ET AL.

MMPs and myocardial infarct. Studies accomplished on the remodeling process following MI have detected higher MMP-1, MMP-2, MMP-3 and MMP-9 expression and activity in human, rat and porcine hearts [66– 70]. Up-regulated MMP activity after infarction suggests that MMPs participate in the cardiac repair process, so these enzymes might play some roles in infarction healing, including early ECM degradation, cell migration (inflammatory cells, fibroblasts), angiogenesis, remodelling of newly synthesized connective tissue and regulation of growth factor activities [2,8]. As discussed by Spinale et al. [3], MMPs assume roles in both the healing process after myocardial injury [71–78] and the adverse remodelling that follows MI [37,79–89]. In the early period after MI (first 72 hr), a wound healing response occurs with appropriate cytokines amplification, inflammatory cells influx and fibroblasts proliferation/transdifferentiation to a myofibroblast phenotype [90–94]. Inflammatory cells and myofibroblasts release some MMPs that can facilitate proteolysis of ECM components, to form nascent wound. Studies performed with transgenic constructs support a relationship between MMP induction and activation after MI [72,73]. Transgenic deletion of MMP-9, MMP-2 or MMP-7 in murine constructs of MI alters the post-MI remodelling process in particular ways [71,74–76]. Molecular imaging approaches in murine models have provided a temporal map of MMP induction and activation within LV during the remodelling process [77,88]. MMP-2 and MMP-9 promoter activation increases within the MI and border zones soon after MI, but higher MMP-2 promoter activation also occurs within the viable remote region in addition to the atrium [77,88]. These studies have cast light on the MMP types that may cause LV remodelling as well as on the regional and temporal aspects of post-MI MMP induction. Data suggest that certain MMP types, such as MMP-1, may not contribute to the adverse LV remodeling process post-MI [88,95]. Studies have demonstrated that global MMP inhibition is unnecessary and that targeting a potential subset of MMP types may be effective [96]. Past studies have suggested that pharmacological MMP inhibition soon after MI is not mandatory; indeed, it can be commenced after the initial wound healing period is complete (first 72 hr) [3]. Studies using MMP inhibitors and genetically modified mice have evidenced the MMP function during LV healing and remodeling following MI. Rohde et al. [84] showed that in vivo MMP inhibition attenuates early LV dilation 4 days after experimental MI in mice. Matrix metalloproteinase inhibitors can be classified as specific or non-specific. Non-specific inhibitors like batimastat, marimastat, GM-6001 (ilomastat or gelardin), PD-166793 and ONO-4817 act by chelating the Zn2+ ion and have found application in many models of the disease [97]. Moreover, the peptides containing the HWGF motif, CRRHWGFEFC and CTTHWGFTLC are selective MMP-9 and MMP-2 inhibitors, respectively [98,99], but their clinical use is uncertain because they are susceptible to proteolysis inside the body. Chemically modified tetracyclines (CMTs) also inhibit MMP activity [100]. CMT-3 suppresses MMP-2 and -9 activities

MiniReview

along with collagenase activity, to improve pathological cardiovascular remodeling. In fact, CMT-3 has been administered to human beings in clinical trials [101]. A common tetracycline antibiotic, doxycycline, has been used as MMP inhibitor in heart failure patients. It inhibits several MMPs and improves ischaemia/reperfusion injury at the onset of MI. Pre-treatment with sub-antimicrobial dose doxycycline (SDD) 2 days prior to MI reduces cardiac dilation and enhances endothelial function in rats [102]. Periostat (a commercial name for doxycycline formulated for periodontal disease) is the only MMP inhibitor that has received approval for clinical use [103]. Brown et al. [104] reported that administration of SDD (two doses of 20 mg/day) to patients with coronary artery disease for months decreased C-reactive protein (CRP) by 46% in the group that received SDD (p = 0.007), but not in the placebo group. Meanwhile, interleukin-6 (IL-6) concentrations went down in the SDD group, but not in the placebo group. Additionally, MMP-9 activity diminished by 50% in the SDD group, suggesting that SDD exerts beneficial effects on inflammation, to probably promote plaque stability [104].

MMPs and atherosclerosis. In atherosclerosis, the MMPs play a part in vascular smooth muscle cell migration and neointima formation after localized vascular injury [105–107]. Active MMPs present in the atherosclerotic lesions may contribute to plaque destabilization by degrading ECM components [108]. Atherosclerotic human vessels display increased MMP-2, MMP-3 and MMP-9 levels as compared with healthy human vessels [80,109]. Studies using knockout mice have suggested that MMP-2, MMP-8, MMP-9 and MMP-12 are implicated in plaque development and burden. MMP-8 [110] and MMP-2 [111] knockout mice with ApoE / atherosclerosis develop significantly reduced atherosclerosis as compared with ApoE / mice. After temporary carotid artery ligation, MMP-9 deficiency also leads to significantly diminished intimal hyperplasia in ApoE / mice [112]. Studies on MMP-12 showed that this MMP promotes lesion initiation and progression in rabbits [113] and that MMP-12 knockout mice with Apo / background have smaller lesions in the brachiocephalic artery without altered lesion growth in aorta [62]. In contrast, genetic MMP-11 [106] or MMP-7 [114] deletion in ApoE / mice does not modify the vascular phenotype or the atherosclerotic lesion, respectively. Additionally, MMP-11 deletion in ApoE / knockout mice enhances neointimal formation after vascular injury as compared with wildtype mice [106]. Matrix metalloproteinases assume a complex role in angiogenesis regulation. In atherosclerosis, angiogenesis occurs by neovascularization that starts in vasa vasorum of the adventicia and then extends towards the intima layer [115,116]. This process may be a response to tissue hypoxia, in an attempt to decrease ischaemia in the tissue of the atherosclerotic lesion. Still concerning atherosclerosis, angiogenesis is also associated with increased atherosclerotic lesion and higher atherosclerotic

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)

MiniReview

MATRIX METALLOPROTEINASES ARE INVOLVED IN CARDIOVASCULAR DISEASES

plaque instability [117,118]. Indeed, treatment of ApoE / mice with angiostatin (inhibitor of angiogenesis) retards lesion progression [119]. Unstable plaques exist within vasa vasorum and apoptotic macrophages, suggesting that these microvessels play essential roles in atherosclerotic lesion progression and instability [120,121]. Matrix metalloproteinase-14 regulates endothelial cell migration and lumen formation within the ECM by controlling capillary sprouting, collagenolysis and endothelial cell invasion [122,123]. MMP-7 is an important regulator in angiogenesis, because it promotes endothelial proliferation [124]. On the other hand, MMP-9 takes on both a pro- and an antiangiogenic role [125,126]. Fang et al. [127] reported fewer endothelial cells or plaque angiogenesis in the atherosclerotic plaques of MMP-8 knockout mice, indicating that MMP-8 is important in ECM angiogenesis in vitro, ex vivo and in vivo. In recent years, countless researchers have investigated how MMP-2 functions in the vascular system. An in vivo study has described stable and significant remodelling of small mesenteric arteries with increased MMP-2 activity after 7 and 15 days of hypertension induced by L-NAME (a non-specific inhibitor of oxide nitric synthase). After 28 days, the MMP-2 activity drops to baseline levels [128]. In this same work, the authors concluded that the eutrophic remodelling of small arteries is associated with varying expression of several proteins involved in cell–matrix interactions.

MMPs and hypertension. Matrix metalloproteinases play an essential role in hypertension [129]. Our group has reported elevated MMP-2 levels in the thoracic aortas [130–138] and heart [139,140] of two-kid-

305

ney, one-clip (2K1C) hypertensive rats. A time course study testing MMP-2 involvement in the vascular alterations in the aorta tissue of 2K1C rats revealed structural changes, augmented MMP-2 levels, gelatinolytic activity, oxidative stress and endothelial dysfuncition in the early phase (2 weeks) of hypertension [134], suggesting that treatment with doxycycline [130,135] and tempol [131] after the first 2 weeks still reverts the alterations in 2K1C rats. Other drugs like dihydropyridines [136], spironolactone and hydrochlorothiazide [133] also prevent vascular remodelling and diminish the MMP-2 levels and gelatinolytic activity in the aortas of 2K1C rats. Many papers have suggested that drugs used in the therapy of cardiovascular diseases may affect MMP activity. However, recent investigations have shown that the MMP inhibitory effect of antihypertensive drugs is more likely a consequence of reduced oxidative stress and lower blood pressure rather than a result of direct MMP inhibition by these drugs. This is the situation verified for the calcium channel blockers nifedipine (0, 1, 10 and 100 mM) and metoprolol (0, 1, 10 and 100 mM) [141] as well as for the diuretics hydrochlorothiazide (0, 1, 10 and 100 lM) and spironolactone (SPRL) (0, 1, 10 and 100 lM) [133]. Figure 1 schematically illustrates those effects. As for treatment of hypertensive patients with enalapril, it significantly lowers systolic and diastolic blood pressure after four and 8 weeks and does not affect MMP activity in the plasma [142]. A series of studies [143–146] claimed that captopril and lisinopril exhibit inhibitory activity. An in vitro study by Reinhardt et al. [145] described the inhibitory activity of captopril, lisinopril and ramiprilate at millimolar concentrations on the basis of zymograms. Since then, zymogram-based other studies have continued to find gelatinases (MMP-2 and MMP-9) inhibition at millimolar concentrations

Fig. 1. Biochemical and functional modifications that occur in aorta and heart of 2K1C animals treated or not with drugs. Heart and aorta of 2K1C-hypertensive animals show increased oxidative stress, and increased matrix metalloproteinase-2 (MMP-2) activity and protein expression. This is associated with cardiac remodelling and endothelial and cardiac dysfunction. Increased oxidative stress also leads directly to cardiac and endothelial dysfunction. The treatment with non-selective inhibitors of MMPs (doxycycline – doxy), reactive oxygen species scavenger (Tempol) and antihypertensive drugs (AHD: spironolactone, hydrochlorothiazide, nifedipine and metoprolol) decreased oxidative stress, activity and expression of MMP-2, reverted the cardiac and vascular remodelling and improved endothelial and cardiac function. Doxy also decreased MMP-2 activity. *AHD: antihypertensive drugs were only tested in thoracic aorta isolated from 2K1C animals.

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)

306

ALINE AZEVEDO ET AL.

only. Indeed, as suggested by Reinhardt et al. [145], the drug has difficulty entering the gel matrix. Most importantly, in the clinical setting, these drugs do not emerge at millimolar levels in the plasma or tissues. Using highly purified MMP-2 to conduct both zymograms and fluorimetric assays, Kuntze et al. [147] have shown that the pH of the solution containing the drugs may drop, which is most likely the reason why a series of studies in the past found that captopril inhibited gelatinases. MMP-2 inhibition by captopril and lisinopril only takes place at millimolar levels. Thus, direct MMP inhibition by ACE inhibitors is probably only a minor concern for the in vivo situation [147]. On the other hand, many other studies have shown that in vitro incubation with captopril and lisinopril significantly reduces MMP-2 activity and that in vivo treatment with these drugs fails to identify a reduction in the accessible pool of MMP-2 protein [146]. Moreover, it has been demonstrated that captopril and lisinopril at 20–40 and 200–400 mM, respectively, lower the gelatinolytic activity in a dose-dependent manner [144]. Chronic exposure to lead, a toxic metal, is associated with hypertension in animals [148–150] and human beings [151– 153]. Rats that were exposed to lead and treated with doxycycline for 8 weeks showed attenuated MMP-2 expression (mRNA, protein and activity) and reduced systolic blood pressure. In addition, the treatment with doxycycline also prevented lead-induced increases in the MMP-2/TIMP-2 mRNA ratio. These results suggested that MMP-2 could play a role in lead-induced increases in blood pressure [154]. Studies have also found increased MMP-2 activity in the thoracic aorta of diabetic rats [155,156]; treatment with doxycycline reverts the endothelial dysfunction in these animals [156,157]. Hence, increased MMP-2 activity may account for the lower vascular relaxation induced by acetylcholine. Zeydanli et al. [157] showed that MMP-2 activity and expression do not change in diabetic rats, but doxycycline decreases lipid peroxidation and cellular glutathione level measured in the plasma and improves endothelial dysfunction. Treatment of animals with doxycycline diminishes the oxidative stress, which may account for improved vascular relaxation [130,135,155–158]. Castro et al. [158] described that treatment of 2K1C rats with doxycycline decreases oxidative stress while increasing nitric oxide (NO) bioavailability in endothelial cells; it also improves endothelium-dependent vascular relaxation induced by A23187 [158]. Another study reported that treatment of spontaneously hypertensive rats (SHRs) with doxycycline reduces oxidative stress and blood pressure [159]. ROS and NO interaction can decrease NO bioavailability and generate peroxynitrite; ROS may also affect the bioavailability of the cofactors of NO synthase, leading to uncoupled NOS, which in turn starts to produce O2 rather than NO [160–164]. Spontaneously hypertensive rats have elevated proteolytic activity in the heart, aorta [159] and plasma [165–168]. Tran et al. [168] showed that SHRs aged between 12 and 18 weeks present increased MMP-2, MMP-9 and MMP-7 activity in the plasma. In a study that evaluated the MMP-2 and MMP-9 lev-

MiniReview

els in the plasma of 8-week-old SHRs and untreated hypertensive patients (with no other diseases) and compared them with data obtained for control animals and normotensive patients, the authors concluded that hypertensive patients and SHR display higher MMP-9 levels as compared with normotensive patients and control animals. These parallel alterations in clinical hypertension and in SHR suggest an important role for MMPs in hypertension [167]. Matrix metalloproteinases cleave the beta-2 adrenergic receptor, which appears to be a potentially important mechanistic link between higher MMP expression and activity and increased arteriolar tone in SHR [169]. Treatment with acid ammonium pyrrolidine dithiocarbamate (DPTC) blocks this process, suggesting that NFkB is responsible for increased MMP activity and cleavage of the beta-2 adrenergic receptor [170]. However, which MMP is involved in this process remains unclear. Matrix metalloproteinase-9 knockout mice present increased vasodilation (endothelium-dependent) of resistance arteries and elevated endothelial nitric oxide synthase (eNOS) [171]. In fact, vasoconstriction of arterioles and venules rapidly takes place in the microvessels of Wistar rats after injection of MMP-7 and MMP-9 (alone or in association) [169]. The mechanism involved in vasoconstriction by MMP-7 depends on the epidermal growth receptor (EGHF), which is important to maintain the vascular tone [172]. In vitro studies have indicated that MMP-2 could play an important role in blood pressure regulation by cleaving big endothelin-1 (Big ET-1) to a potent vasoconstrictor, endothelin-1 (ET 1–32) [173]. In vitro MMP-2 could also cleave the vasodilator peptide CGRP (calcitonin gene-related peptide), generating degradation products with lower vasodilating action [174]. Also, in vitro MMP-2 could act on adrenomedullin, producing metabolites with lower vasodilating potential and which actually act as vasoconstrictors [175]. Playing the role of a coadjuvant, MMP-2 released by thrombin action could cleave Big ET-1, to give 1–32 ET. In arteries with endothelium, 1–32 ET forms NO and prostaglandin 2 (PGI2), which culminates in vascular relaxation; in endothelium-denuded arteries, vasoconstriction should occur [176].

MMPs and cardiomyopathies. A few studies have shown that MMP-2 alone accounts for severe morphological changes and functional loss in the heart. Matsusaka et al. [177] reported that the absence of MMP-2 protects the heart from injury-induced increase in the ventricular afterload. A time course study in 2K1C rats showed increased ROS levels, TGF beta expression and MMP-2 activity and expression 15–75 days after arterial clipping. Left ventricular and septum wall thickening and cardiomyocyte hypertrophy were also detected in this situation [178]. Treatment of 2K1C animals with tempol [139] and doxycycline [140] reversed cardiac alterations. Increased MMP-2 in the heart of the transgenic model augments the infarction areas and diminishes the contractile

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)

MiniReview

MATRIX METALLOPROTEINASES ARE INVOLVED IN CARDIOVASCULAR DISEASES

function in the hearts of animals subjected to ischaemia/reperfusion [179]. These transgenic mice express active MMP-2 (muted) driven by the myosin heavy chain promoter. The prodomain-muted MMP-2 couples to a c-myc epitope tag, to differentiate the endogenous MMP-2 [180]. Active MMP-2 expression in the heart of transgenic mice results in cardiac functional loss and morphological alterations, marked ventricular remodeling with myocyte hypertrophy, myofilaments lysis, sarcomeres destruction and cardiac fibroblasts proliferation [180]. Increased MMP-2 in the heart also degenerates the heart valves after 12–24 weeks of life [181]. More recently, Lovett et al. [40] verified that oxidative stress in cardiac myocytes induces a novel intracellular (mitochondrial) MMP-2 isoform [40]. This form has an N-terminal truncation (the first methionine corresponds to methionine 77 of the full-length MMP-2) and is shorter (65 kDa). Because this isoform loses part of the propeptide, it probably becomes active at the moment it is translated, so this shorter intracellular MMP-2 form may contribute to the structural and functional cardiac damage that occurs in many cardiovascular diseases leading to heart failure. Moreover, MMP-2 found in sarcomers cleaves many structural cardiac proteins, like troponin I [45], myosin light chain 1 [43], a-actinin [44] and titin [42] contributing to decreased cardiac function. Figure 2 schematically illustrates the activity of MMP-2 on heart proteins and function. Dahi et al. [60] verified that lesions emerge in the coronary arteries during the late period (when higher MMP-2 levels

307

exist in the vessels and begin to decrease in cardiomyocytes), which attests to the critical role of increased MMP-2 expression. These authors described that the elastin of arteries loses integrity, fibrous tissue deposits in the adventitia and inflammatory infiltrate increases. The kidney is essential for blood pressure control; after the first week of life, MMP-2 expression by transgenesis (mice) in the epithelial cells of proximal tubules leads to severe structural disorganization of the kidney parenchyma, culminating in changes in the tubular basement membrane, epithelial-mesenchymal transition, tubular atrophy, tubular fibrosis and renal function loss [182]. When it is active in specific organs that are essential for cardiovascular function (heart and kidney), MMP-2 alone is sufficient to induce changes in animals that mimic well-known clinical conditions. This information is very important when one considers (1) studies that have demonstrated the role increased MMP-2 expression plays in the heart and vessels during various cardiac diseases and (2) investigations showing that decreased MMP-2 activity reverses cardiovascular conditions such as hypertension. Understanding how one protease alone (MMP-2) acts in some cardiovascular diseases opens new therapeutic possibilities. Indeed, Periostat has been clinically used in the United States to treat periodontitis, a disease of the gums and teeth [103]. MMPs and periodontitis. Periodontitis is a chronic disease that is highly associated with cardiovascular diseases [183]; this association is important to

Fig. 2. Active matrix metalloproteinase-2 (MMP-2) involvement in cardiac dysfunction in cardiovascular diseases. Inflammatory cells, reactive oxygen species and fibroblast proliferation lead to higher activity and/or increased protein expression of MMP-2 in pathological conditions, including ischaemia/reperfusion, ventricular afterload, 2K1C hypertension and myocardial infarct. The higher level/activity of MMP-2 leads to cardiac dysfunctions (as indicated in the figure) by several ways, including alteration of cardiac cells phenotype (cardiomyocyte hypertrophy) and cardiac remodelling (ventricular dilatation) or degradation of contractile and structural proteins (troponin I, myosin light chain 1, a-actinin and titin) and heart valve degeneration.

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)

ALINE AZEVEDO ET AL.

308

clinically manage cardiovascular conditions [184]. MMPs have already been recognized to be essential to the progress of periodontitis; they are the most important class of proteases to degrade the collagen fibres that constitute the periodontal ligament and maintain the tooth attached to the bone. MMPs are also crucial to the inflammatory response that recruits inflammatory cells to the inflammation site. The FDA has approved doxycycline to treat periodontal disease. This antibiotics reduces some inflammatory markers that are important for cardiovascular events such as cell adhesion molecules (ICAM and VCAM), cytokines, tumour necrosis factor alpha (TNF), CD 40, IL-6, fibrinogen, serum amyloid A (SAA), CRP and lipoprotein phospholipase A2 in the plasma [185]. Treating periodontal disease decreases IL-6 and CRP plasma levels [186,187], reduces the carotid intima diameter the after 6 weeks [188] and diminishes the MMP-8 and MMP-9 plasma concentrations [186]. Moreover, periodontitis has been linked to atherosclerotic development in mice models [105,189,190]. Treatment with doxycycline reduced pro-inflammatory cytokines and MMP-9 plasma levels and diminished atherosclerosis in apoE+/ inoculated with periodontal pathogenic bacteria [191]. Interestingly, MMP-9 has been described as a possible biomarker of cardiovascular events [49,107,192–197]. Therefore, increased MMP activity in the plasma and in organs (and not only an increased pro-inflammatory state) may be the link between periodontitis and cardiovascular diseases. Conclusions Advances have been made towards understanding of the biochemical and structural aspects of MMPs, including their activation and catalytic mechanisms, substrates specificity and mechanism of inhibition. MMPs are involved in many diseases. Using different models, researchers have gathered substantial evidence that MMPs mediate the progress of various cardiovascular diseases. MMP-2 is one of the MMPs that has been extensively described in the heart and in normal and diseased vessels. Our group has shown increased MMP-2 levels in hypertrophic 2K1C aortas of hypertensive rats; administration of a non-specific MMP inhibitor, doxycycline, significantly reduces the blood pressure and completely reverses the structural changes in the vessels. A link between cardiovascular diseases and periodontitis also exists; it may be mediated by MMPs, as both conditions involve increased MMP expression. Further studies on the involvement of specific MMPs in disease models may help elucidate the roles of MMP and aid discovery of new drug targets to treat cardiovascular diseases. Acknowledgements This work was supported by CAPES (Brasilia, Brazil) and FAPESP (State of Sao Paulo Research Foundation, SP, Brazil). A. Azevedo received a fellowship from CAPES (Grant Number 02764/09-1). References 1 Nagase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res 2006;69:562–73.

MiniReview

2 Nagase H, Woessner F Jr. Matrix metalloproteinases. J Biol Chem 1999;274:21491–4. 3 Spinale FG, Janicki JS, Zile MR. Membrane-associated matrix proteolysis and heart failure. Circ Res 2013;112:195–208. 4 Siefert SA, Sarkar R. Matrix metalloproteinases in vascular physiology and disease. Vascular 2012;20:210–6. 5 Hooper NM. Families of zinc metalloproteases. FEBS Lett 1994;354:1–6. 6 Gross J, Lapiere CM. Collagenolytic activity in amphibian tissues: a tissue culture assay. Proc Natl Acad Sci USA 1962;48:1014–22. 7 Woolley DE, Roberts DR, Evanson JM. Inhibition of human collagenase activity by a small molecular weight serum protein. Biochem Biophys Res Commun 1975;66:747–54. 8 Page-McCaw A, Ewald AJ, Werb Z. Matrix metalloproteinases and the regulation of tissue remodelling. Nat Rev Mol Cell Biol 2007;8:221–33. 9 Wada K, Sato H, Kinoh H, Kajita M, Yamamoto H, Seiki M. Cloning of three Caenorhabditis elegans genes potentially encoding novel matrix metalloproteinases. Gene 1998;211:57–62. 10 Lepage T, Gache C. Early expression of a collagenase-like hatching enzyme gene in the sea urchin embryo. EMBO J 1990;9:3003–12. 11 Leontovich AA, Zhang J, Shimokawa K, Nagase H, Sarras MP Jr. A novel hydra matrix metalloproteinase (HMMP) functions in extracellular matrix degradation, morphogenesis and the maintenance of differentiated cells in the foot process. Development 2000;127:907–20. 12 Massova I, Kotra LP, Fridman R, Mobashery S. Matrix metalloproteinases: structures, evolution, and diversification. FASEB J 1998;12:1075–95. 13 Van Wart HE, Birkedal-Hansen H. The cysteine switch: a principle of regulation of metalloproteinase activity with potential applicability to the entire matrix metalloproteinase gene family. Proc Natl Acad Sci USA 1990;87:5578–82. 14 Bode W, Gomis-Ruth FX, Stockler W. Astacins, serralysins, snake venom and matrix metalloproteinases exhibit identical zinc-binding environments (HEXXHXXGXXH and Met-turn) and topologies and should be grouped into a common family, the ‘metzincins’. FEBS Lett 1993;331:134–40. 15 Woessner JF Jr. Matrix metalloproteinases and their inhibitors in connective tissue remodeling. FASEB J 1991;5:2145–54. 16 Murphy G, Stanton H, Cowell S, Butler G, Knauper V, Atkinson S et al. Mechanisms for pro matrix metalloproteinase activation. APMIS 1999;107:38–44. 17 Knauper V, Will H, Lopez-Otin C, Smith B, Atkinson SJ, Stanton H et al. Cellular mechanisms for human procollagenase-3 (MMP-13) activation. Evidence that MT1-MMP (MMP-14) and gelatinase a (MMP-2) are able to generate active enzyme. J Biol Chem 1996;271:17124–31. 18 Sato H, Takino T, Okada Y, Cao J, Shinagawa A, Yamamoto E et al. A matrix metalloproteinase expressed on the surface of invasive tumour cells. Nature 1994;370:61–5. 19 Butler GS, Butler MJ, Atkinson SJ, Will H, Tamura T, Schade van Westrum S et al. The TIMP2 membrane type 1 metalloproteinase “receptor” regulates the concentration and efficient activation of progelatinase A. A kinetic study. J Biol Chem 1998;273:871–80. 20 Strongin AY, Collier I, Bannikov G, Marmer BL, Grant GA, Goldberg GI. Mechanism of cell surface activation of 72-kDa type IV collagenase. Isolation of the activated form of the membrane metalloprotease. J Biol Chem 1995;270:5331–8. 21 Wang Z, Juttermann R, Soloway PD. TIMP-2 is required for efficient activation of proMMP-2 in vivo. J Biol Chem 2000;275:26411–5. 22 Worley JR, Thompkins PB, Lee MH, Hutton M, Soloway P, Edwards DR et al. Sequence motifs of tissue inhibitor of metal-

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)

MiniReview

23

24

25

26

27

28 29

30

31

32

33

34

35

36

37

38

MATRIX METALLOPROTEINASES ARE INVOLVED IN CARDIOVASCULAR DISEASES

loproteinases 2 (TIMP-2) determining progelatinase A (proMMP-2) binding and activation by membrane-type metalloproteinase 1 (MT1-MMP). Biochem J 2003;372:799–809. Kinoshita T, Sato H, Okada A, Ohuchi E, Imai K, Okada Y et al. TIMP-2 promotes activation of progelatinase A by membrane-type 1 matrix metalloproteinase immobilized on agarose beads. J Biol Chem 1998;273:16098–103. Itoh Y, Takamura A, Ito N, Maru Y, Sato H, Suenaga N et al. Homophilic complex formation of MT1-MMP facilitates proMMP-2 activation on the cell surface and promotes tumor cell invasion. EMBO J 2001;20:4782–93. Atkinson SJ, Crabbe T, Cowell S, Ward RV, Butler MJ, Sato H et al. Intermolecular autolytic cleavage can contribute to the activation of progelatinase A by cell membranes. J Biol Chem 1995;270:30479–85. Viappiani S, Nicolescu AC, Holt A, Sawicki G, Crawford BD, Leon H et al. Activation and modulation of 72 kDa matrix metalloproteinase-2 by peroxynitrite and glutathione. Biochem Pharmacol 2009;77:826–34. Okamoto T, Akaike T, Sawa T, Miyamoto Y, van der Vliet A, Maeda H. Activation of matrix metalloproteinases by peroxynitrite-induced protein S-glutathiolation via disulfide S-oxide formation. J Biol Chem 2001;276:29596–602. Bode W. A helping hand for collagenases: the haemopexin-like domain. Structure 1995;3:527–30. Allan JA, Docherty AJ, Barker PJ, Huskisson NS, Reynolds JJ, Murphy G. Binding of gelatinases A and B to type-I collagen and other matrix components. Biochem J 1995;309(Pt 1):299– 306. Steffensen B, Wallon UM, Overall CM. Extracellular matrix binding properties of recombinant fibronectin type II-like modules of human 72-kDa gelatinase/type IV collagenase. High affinity binding to native type I collagen but not native type IV collagen. J Biol Chem 1995;270:11555–66. Goncalves AN, Meschiari CA, Stetler-Stevenson WG, Nonato MC, Alves CP, Espreafico EM et al. Expression of soluble and functional full-length human matrix metalloproteinase-2 in Escherichia coli. J Biotechnol 2012;157:20–4. Benbow U, Brinckerhoff CE. The AP-1 site and MMP gene regulation: what is all the fuss about? Matrix Biol 1997;15: 519–26. Bergman MR, Cheng S, Honbo N, Piacentini L, Karliner JS, Lovett DH. A functional activating protein 1 (AP-1) site regulates matrix metalloproteinase 2 (MMP-2) transcription by cardiac cells through interactions with JunB-Fra1 and JunB-FosB heterodimers. Biochem J 2003;369:485–96. Alfonso-Jaume MA, Bergman MR, Mahimkar R, Cheng S, Jin ZQ, Karliner JS et al. Cardiac ischemia-reperfusion injury induces matrix metalloproteinase-2 expression through the AP-1 components FosB and JunB. Am J Physiol Heart Circ Physiol 2006;291:H1838–46. Sariahmetoglu M, Crawford BD, Leon H, Sawicka J, Li L, Ballermann BJ et al. Regulation of matrix metalloproteinase-2 (MMP-2) activity by phosphorylation. FASEB J 2007;21:2486–95. Gomez DE, Alonso DF, Yoshiji H, Thorgeirsson UP. Tissue inhibitors of metalloproteinases: structure, regulation and biological functions. Eur J Cell Biol 1997;74:111–22. Webb CS, Bonnema DD, Ahmed SH, Leonardi AH, McClure CD, Clark LL et al. Specific temporal profile of matrix metalloproteinase release occurs in patients after myocardial infarction: relation to left ventricular remodeling. Circulation 2006;114:1020–7. Ahmed SH, Clark LL, Pennington WR, Webb CS, Bonnema DD, Leonardi AH et al. Matrix metalloproteinases/tissue inhibitors of metalloproteinases: relationship between changes in proteolytic determinants of matrix composition and structural, functional,

39

40

41

42

43

44

45

46

47

48

49

50

51

52

53

309

and clinical manifestations of hypertensive heart disease. Circulation 2006;113:2089–96. Si-Tayeb K, Monvoisin A, Mazzocco C, Lepreux S, Decossas M, Cubel G et al. Matrix metalloproteinase 3 is present in the cell nucleus and is involved in apoptosis. Am J Pathol 2006;169:1390–401. Lovett DH, Mahimkar R, Raffai RL, Cape L, Maklashina E, Cecchini G et al. A novel intracellular isoform of matrix metalloproteinase-2 induced by oxidative stress activates innate immunity. PLoS One 2012;7:e34177. Kwan JA, Schulze CJ, Wang W, Leon H, Sariahmetoglu M, Sung M et al. Matrix metalloproteinase-2 (MMP-2) is present in the nucleus of cardiac myocytes and is capable of cleaving poly (ADP-ribose) polymerase (PARP) in vitro. FASEB J 2004;18:690–2. Ali MA, Cho WJ, Hudson B, Kassiri Z, Granzier H, Schulz R. Titin is a target of matrix metalloproteinase-2: implications in myocardial ischemia/reperfusion injury. Circulation 2010;122:2039–47. Sawicki G, Leon H, Sawicka J, Sariahmetoglu M, Schulze CJ, Scott PG et al. Degradation of myosin light chain in isolated rat hearts subjected to ischemia-reperfusion injury: a new intracellular target for matrix metalloproteinase-2. Circulation 2005;112:544–52. Sung MM, Schulz CG, Wang W, Sawicki G, Bautista-Lopez NL, Schulz R. Matrix metalloproteinase-2 degrades the cytoskeletal protein alpha-actinin in peroxynitrite mediated myocardial injury. J Mol Cell Cardiol 2007;43:429–36. Wang W, Schulze CJ, Suarez-Pinzon WL, Dyck JR, Sawicki G, Schulz R. Intracellular action of matrix metalloproteinase-2 accounts for acute myocardial ischemia and reperfusion injury. Circulation 2002;106:1543–9. Morgan AR, Rerkasem K, Gallagher PJ, Zhang B, Morris GE, Calder PC et al. Differences in matrix metalloproteinase-1 and matrix metalloproteinase-12 transcript levels among carotid atherosclerotic plaques with different histopathological characteristics. Stroke 2004;35:1310–5. Nikkari ST, Geary RL, Hatsukami T, Ferguson M, Forough R, Alpers CE et al. Expression of collagen, interstitial collagenase, and tissue inhibitor of metalloproteinases-1 in restenosis after carotid endarterectomy. Am J Pathol 1996;148:777–83. Shah PK, Falk E, Badimon JJ, Fernandez-Ortiz A, Mailhac A, Villareal-Levy G et al. Human monocyte-derived macrophages induce collagen breakdown in fibrous caps of atherosclerotic plaques. Potential role of matrix-degrading metalloproteinases and implications for plaque rupture. Circulation 1995;92: 1565–9. Zeng B, Prasan A, Fung KC, Solanki V, Bruce D, Freedman SB et al. Elevated circulating levels of matrix metalloproteinase-9 and -2 in patients with symptomatic coronary artery disease. Intern Med J 2005;35:331–5. Brophy CM, Marks WH, Reilly JM, Tilson MD. Decreased tissue inhibitor of metalloproteinases (TIMP) in abdominal aortic aneurysm tissue: a preliminary report. J Surg Res 1991;50:653–7. Thompson RW, Holmes DR, Mertens RA, Liao S, Botney MD, Mecham RP et al. Production and localization of 92-kilodalton gelatinase in abdominal aortic aneurysms. An elastolytic metalloproteinase expressed by aneurysm-infiltrating macrophages. J Clin Invest 1995;96:318–26. Celentano DC, Frishman WH. Matrix metalloproteinases and coronary artery disease: a novel therapeutic target. J Clin Pharmacol 1997;37:991–1000. Galis ZS, Sukhova GK, Lark MW, Libby P. Increased expression of matrix metalloproteinases and matrix degrading activity in vulnerable regions of human atherosclerotic plaques. J Clin Invest 1994;94:2493–503.

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)

310

ALINE AZEVEDO ET AL.

54 Halpert I, Sires UI, Roby JD, Potter-Perigo S, Wight TN, Shapiro SD et al. Matrilysin is expressed by lipid-laden macrophages at sites of potential rupture in atherosclerotic lesions and localizes to areas of versican deposition, a proteoglycan substrate for the enzyme. Proc Natl Acad Sci USA 1996;93:9748–53. 55 Knox JB, Sukhova GK, Whittemore AD, Libby P. Evidence for altered balance between matrix metalloproteinases and their inhibitors in human aortic diseases. Circulation 1997;95:205– 12. 56 Mata KM, Prudente PS, Rocha FS, Prado CM, Floriano EM, Elias J Jr et al. Combining two potential causes of metalloproteinase secretion causes abdominal aortic aneurysms in rats: a new experimental model. Int J Exp Pathol 2011;92:26–39. 57 Wilson WR, Schwalbe EC, Jones JL, Bell PR, Thompson MM. Matrix metalloproteinase 8 (neutrophil collagenase) in the pathogenesis of abdominal aortic aneurysm. Br J Surg 2005;92:828– 33. 58 Basu R, Fan D, Kandalam V, Lee J, Das SK, Wang X et al. Loss of Timp3 gene leads to abdominal aortic aneurysm formation in response to angiotensin II. J Biol Chem 2012;287:44083– 96. 59 Xiong W, Knispel R, Mactaggart J, Baxter BT. Effects of tissue inhibitor of metalloproteinase 2 deficiency on aneurysm formation. J Vasc Surg 2006;44:1061–6. 60 Dahi S, Karliner JS, Sarkar R, Lovett DH. Transgenic expression of matrix metalloproteinase-2 induces coronary artery ectasia. Int J Exp Pathol 2011;92:50–6. 61 Longo GM, Buda SJ, Fiotta N, Xiong W, Griener T, Shapiro S et al. MMP-12 has a role in abdominal aortic aneurysms in mice. Surgery 2005;137:457–62. 62 Luttun A, Lutgens E, Manderveld A, Maris K, Collen D, Carmeliet P et al. Loss of matrix metalloproteinase-9 or matrix metalloproteinase-12 protects apolipoprotein E-deficient mice against atherosclerotic media destruction but differentially affects plaque growth. Circulation 2004;109:1408–14. 63 Alleva R, Nasole E, Di Donato F, Borghi B, Neuzil J, Tomasetti M. Alpha-Lipoic acid supplementation inhibits oxidative damage, accelerating chronic wound healing in patients undergoing hyperbaric oxygen therapy. Biochem Biophys Res Commun 2005;333:404–10. 64 Mustoe T. Understanding chronic wounds: a unifying hypothesis on their pathogenesis and implications for therapy. Am J Surg 2004;187:65S–70S. 65 Mishra PK, Tyagi N, Sen U, Joshua IG, Tyagi SC. Synergism in hyperhomocysteinemia and diabetes: role of PPAR gamma and tempol. Cardiovasc Diabetol 2010;9:49. 66 Carlyle WC, Jacobson AW, Judd DL, Tian B, Chu C, Hauer KM et al. Delayed reperfusion alters matrix metalloproteinase activity and fibronectin mRNA expression in the infarct zone of the ligated rat heart. J Mol Cell Cardiol 1997;29:2451–63. 67 Cleutjens JP, Kandala JC, Guarda E, Guntaka RV, Weber KT. Regulation of collagen degradation in the rat myocardium after infarction. J Mol Cell Cardiol 1995;27:1281–92. 68 Danielsen CC, Wiggers H, Andersen HR. Increased amounts of collagenase and gelatinase in porcine myocardium following ischemia and reperfusion. J Mol Cell Cardiol 1998;30:1431–42. 69 Sato S, Ashraf M, Millard RW, Fujiwara H, Schwartz A. Connective tissue changes in early ischemia of porcine myocardium: an ultrastructural study. J Mol Cell Cardiol 1983;15:261–75. 70 Tyagi SC, Kumar SG, Haas SJ, Reddy HK, Voelker DJ, Hayden MR et al. Post-transcriptional regulation of extracellular matrix metalloproteinase in human heart end-stage failure secondary to ischemic cardiomyopathy. J Mol Cell Cardiol 1996;28:1415–28. 71 Ducharme A, Frantz S, Aikawa M, Rabkin E, Lindsey M, Rohde LE et al. Targeted deletion of matrix metalloproteinase-9 attenuates left ventricular enlargement and collagen accumulation after

72

73

74

75

76

77

78

79

80

81

82

83

84

85

86

MiniReview

experimental myocardial infarction. J Clin Invest 2000;106: 55–62. Heymans S, Luttun A, Nuyens D, Theilmeier G, Creemers E, Moons L et al. Inhibition of plasminogen activators or matrix metalloproteinases prevents cardiac rupture but impairs therapeutic angiogenesis and causes cardiac failure. Nat Med 1999;5:1135–42. Ikonomidis JS, Hendrick JW, Parkhurst AM, Herron AR, Escobar PG, Dowdy KB et al. Accelerated LV remodeling after myocardial infarction in TIMP-1-deficient mice: effects of exogenous MMP inhibition. Am J Physiol Heart Circ Physiol 2005;288: H149–58. Lindsey ML, Escobar GP, Dobrucki LW, Goshorn DK, Bouges S, Mingoia JT et al. Matrix metalloproteinase-9 gene deletion facilitates angiogenesis after myocardial infarction. Am J Physiol Heart Circ Physiol 2006;290:H232–9. Lindsey ML, Escobar GP, Mukherjee R, Goshorn DK, Sheats NJ, Bruce JA et al. Matrix metalloproteinase-7 affects connexin43 levels, electrical conduction, and survival after myocardial infarction. Circulation 2006;113:2919–28. Matsumura S, Iwanaga S, Mochizuki S, Okamoto H, Ogawa S, Okada Y. Targeted deletion or pharmacological inhibition of MMP-2 prevents cardiac rupture after myocardial infarction in mice. J Clin Invest 2005;115:599–609. Mukherjee R, Mingoia JT, Bruce JA, Austin JS, Stroud RE, Escobar GP et al. Selective spatiotemporal induction of matrix metalloproteinase-2 and matrix metalloproteinase-9 transcription after myocardial infarction. Am J Physiol Heart Circ Physiol 2006;291:H2216–28. Spinale FG, Escobar GP, Mukherjee R, Zavadzkas JA, Saunders SM, Jeffords LB et al. Cardiac-restricted overexpression of membrane type-1 matrix metalloproteinase in mice: effects on myocardial remodeling with aging. Circ Heart Fail 2009;2:351–60. Elliot S, Catanuto P, Fernandez P, Espinosa-Heidmann D, Karl M, Korach K et al. Subtype specific estrogen receptor action protects against changes in MMP-2 activation in mouse retinal pigmented epithelial cells. Exp Eye Res 2008;86:653–60. Kai H, Ikeda H, Yasukawa H, Kai M, Seki Y, Kuwahara F et al. Peripheral blood levels of matrix metalloproteases-2 and -9 are elevated in patients with acute coronary syndromes. J Am Coll Cardiol 1998;32:368–72. Kelly D, Cockerill G, Ng LL, Thompson M, Khan S, Samani NJ et al. Plasma matrix metalloproteinase-9 and left ventricular remodelling after acute myocardial infarction in man: a prospective cohort study. Eur Heart J 2007;28:711–8. Mukherjee R, Brinsa TA, Dowdy KB, Scott AA, Baskin JM, Deschamps AM et al. Myocardial infarct expansion and matrix metalloproteinase inhibition. Circulation 2003;107:618–25. Nilsson L, Hallen J, Atar D, Jonasson L, Swahn E. Early measurements of plasma matrix metalloproteinase-2 predict infarct size and ventricular dysfunction in ST-elevation myocardial infarction. Heart 2012;98:31–6. Rohde LE, Ducharme A, Arroyo LH, Aikawa M, Sukhova GH, Lopez-Anaya A et al. Matrix metalloproteinase inhibition attenuates early left ventricular enlargement after experimental myocardial infarction in mice. Circulation 1999;99:3063–70. Sahul ZH, Mukherjee R, Song J, McAteer J, Stroud RE, Dione DP et al. Targeted imaging of the spatial and temporal variation of matrix metalloproteinase activity in a porcine model of postinfarct remodeling: relationship to myocardial dysfunction. Circ Cardiovasc Imaging 2011;4:381–91. Spinale FG, Coker ML, Thomas CV, Walker JD, Mukherjee R, Hebbar L. Time-dependent changes in matrix metalloproteinase activity and expression during the progression of congestive heart failure: relation to ventricular and myocyte function. Circ Res 1998;82:482–95.

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)

MiniReview

MATRIX METALLOPROTEINASES ARE INVOLVED IN CARDIOVASCULAR DISEASES

87 Spinale FG, Mukherjee R, Zavadzkas JA, Koval CN, Bouges S, Stroud RE et al. Cardiac restricted overexpression of membrane type-1 matrix metalloproteinase causes adverse myocardial remodeling following myocardial infarction. J Biol Chem 2010;285:30316–27. 88 Su H, Spinale FG, Dobrucki LW, Song J, Hua J, Sweterlitsch S et al. Noninvasive targeted imaging of matrix metalloproteinase activation in a murine model of postinfarction remodeling. Circulation 2005;112:3157–67. 89 Yarbrough WM, Mukherjee R, Escobar GP, Mingoia JT, Sample JA, Hendrick JW et al. Selective targeting and timing of matrix metalloproteinase inhibition in post-myocardial infarction remodeling. Circulation 2003;108:1753–9. 90 Daskalopoulos EP, Janssen BJ, Blankesteijn WM. Myofibroblasts in the infarct area: concepts and challenges. Microsc Microanal 2012;18:35–49. 91 Dobaczewski M, Gonzalez-Quesada C, Frangogiannis NG. The extracellular matrix as a modulator of the inflammatory and reparative response following myocardial infarction. J Mol Cell Cardiol 2010;48:504–11. 92 Frangogiannis NG. Regulation of the inflammatory response in cardiac repair. Circ Res 2012;110:159–73. 93 Nistico P, Bissell MJ, Radisky DC. Epithelial-mesenchymal transition: general principles and pathological relevance with special emphasis on the role of matrix metalloproteinases. Cold Spring Harb Perspect Biol 2012;4(2). pii: a011908. doi: 10.1101/cshpe rspect.a011908. 94 Widgerow AD. Cellular/extracellular matrix cross-talk in scar evolution and control. Wound Repair Regen 2011;19:117–33. 95 King MK, Coker ML, Goldberg A, McElmurray JH III, Gunasinghe HR, Mukherjee R et al. Selective matrix metalloproteinase inhibition with developing heart failure: effects on left ventricular function and structure. Circ Res 2003;92:177–85. 96 Peterson JT. The importance of estimating the therapeutic index in the development of matrix metalloproteinase inhibitors. Cardiovasc Res 2006;69:677–87. 97 Peterson JT. Matrix metalloproteinase inhibitor development and the remodeling of drug discovery. Heart Fail Rev 2004;9:63–79. 98 Koivunen E, Arap W, Valtanen H, Rainisalo A, Medina OP, Heikkila P et al. Tumor targeting with a selective gelatinase inhibitor. Nat Biotechnol 1999;17:768–74. 99 Higashi S, Miyazaki K. Identification of a region of beta-amyloid precursor protein essential for its gelatinase A inhibitory activity. J Biol Chem 2003;278:14020–8. 100 Golub LM, Suomalainen K, Sorsa T. Host modulation with tetracyclines and their chemically modified analogues. Curr Opin Dent 1992;2:80–90. 101 Rudek MA, Figg WD, Dyer V, Dahut W, Turner ML, Steinberg SM et al. Phase I clinical trial of oral COL-3, a matrix metalloproteinase inhibitor, in patients with refractory metastatic cancer. J Clin Oncol 2001;19:584–92. 102 Tessone A, Feinberg MS, Barbash IM, Reich R, Holbova R, Richmann M et al. Effect of matrix metalloproteinase inhibition by doxycycline on myocardial healing and remodeling after myocardial infarction. Cardiovasc Drugs Ther 2005;19:383–90. 103 Novak MJ, Johns LP, Miller RC, Bradshaw MH. Adjunctive benefits of subantimicrobial dose doxycycline in the management of severe, generalized, chronic periodontitis. J Periodontol 2002;73:762–9. 104 Brown DL, Desai KK, Vakili BA, Nouneh C, Lee HM, Golub LM. Clinical and biochemical results of the metalloproteinase inhibition with subantimicrobial doses of doxycycline to prevent acute coronary syndromes (MIDAS) pilot trial. Arterioscler Thromb Vasc Biol 2004;24:733–8. 105 Lalla E, Lamster IB, Hofmann MA, Bucciarelli L, Jerud AP, Tucker S et al. Oral infection with a periodontal pathogen accel-

106

107

108 109

110

111

112

113

114

115

116

117

118

119

120

121

122

311

erates early atherosclerosis in apolipoprotein E-null mice. Arterioscler Thromb Vasc Biol 2003;23:1405–11. Lijnen HR, Van Hoef B, Vanlinthout I, Verstreken M, Rio MC, Collen D. Accelerated neointima formation after vascular injury in mice with stromelysin-3 (MMP-11) gene inactivation. Arterioscler Thromb Vasc Biol 1999;19:2863–70. Soder PO, Meurman JH, Jogestrand T, Nowak J, Soder B. Matrix metalloproteinase-9 and tissue inhibitor of matrix metalloproteinase-1 in blood as markers for early atherosclerosis in subjects with chronic periodontitis. J Periodontal Res 2009;44:452–8. Falk E, Shah PK, Fuster V. Coronary plaque disruption. Circulation 1995;92:657–71. Wu TC, Leu HB, Lin WT, Lin CP, Lin SJ, Chen JW. Plasma matrix metalloproteinase-3 level is an independent prognostic factor in stable coronary artery disease. Eur J Clin Invest 2005;35:537–45. Laxton RC, Hu Y, Duchene J, Zhang F, Zhang Z, Leung KY et al. A role of matrix metalloproteinase-8 in atherosclerosis. Circ Res 2009;105:921–9. Kuzuya M, Nakamura K, Sasaki T, Cheng XW, Itohara S, Iguchi A. Effect of MMP-2 deficiency on atherosclerotic lesion formation in apoE-deficient mice. Arterioscler Thromb Vasc Biol 2006;26:1120–5. Galis ZS, Johnson C, Godin D, Magid R, Shipley JM, Senior RM et al. Targeted disruption of the matrix metalloproteinase-9 gene impairs smooth muscle cell migration and geometrical arterial remodeling. Circ Res 2002;91:852–9. Yamada S, Wang KY, Tanimoto A, Fan J, Shimajiri S, Kitajima S et al. Matrix metalloproteinase 12 accelerates the initiation of atherosclerosis and stimulates the progression of fatty streaks to fibrous plaques in transgenic rabbits. Am J Pathol 2008;172:1419–29. Johnson JL, George SJ, Newby AC, Jackson CL. Divergent effects of matrix metalloproteinases 3, 7, 9, and 12 on atherosclerotic plaque stability in mouse brachiocephalic arteries. Proc Natl Acad Sci USA 2005;102:15575–80. Barger AC, Beeuwkes R III, Lainey LL, Silverman KJ. Hypothesis: vasa vasorum and neovascularization of human coronary arteries. A possible role in the pathophysiology of atherosclerosis. N Engl J Med 1984;310:175–7. Kaiser M, Younge B, Bjornsson J, Goronzy JJ, Weyand CM. Formation of new vasa vasorum in vasculitis. Production of angiogenic cytokines by multinucleated giant cells. Am J Pathol 1999;155:765–74. Doyle B, Caplice N. Plaque neovascularization and antiangiogenic therapy for atherosclerosis. J Am Coll Cardiol 2007;49:2073–80. Kolodgie FD, Narula J, Yuan C, Burke AP, Finn AV, Virmani R. Elimination of neoangiogenesis for plaque stabilization: is there a role for local drug therapy? J Am Coll Cardiol 2007;49:2093–101. Moulton KS, Vakili K, Zurakowski D, Soliman M, Butterfield C, Sylvin E et al. Inhibition of plaque neovascularization reduces macrophage accumulation and progression of advanced atherosclerosis. Proc Natl Acad Sci USA 2003;100:4736–41. Kolodgie FD, Virmani R, Burke AP, Farb A, Weber DK, Kutys R et al. Pathologic assessment of the vulnerable human coronary plaque. Heart 2004;90:1385–91. Virmani R, Kolodgie FD, Burke AP, Finn AV, Gold HK, Tulenko TN et al. Atherosclerotic plaque progression and vulnerability to rupture: angiogenesis as a source of intraplaque hemorrhage. Arterioscler Thromb Vasc Biol 2005;25:2054–61. Chun TH, Sabeh F, Ota I, Murphy H, McDonagh KT, Holmbeck K et al. MT1-MMP-dependent neovessel formation within the confines of the three-dimensional extracellular matrix. J Cell Biol 2004;167:757–67.

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)

312

ALINE AZEVEDO ET AL.

123 Robinet A, Fahem A, Cauchard JH, Huet E, Vincent L, Lorimier S et al. Elastin-derived peptides enhance angiogenesis by promoting endothelial cell migration and tubulogenesis through upregulation of MT1-MMP. J Cell Sci 2005;118:343–56. 124 Huo N, Ichikawa Y, Kamiyama M, Ishikawa T, Hamaguchi Y, Hasegawa S et al. MMP-7 (matrilysin) accelerated growth of human umbilical vein endothelial cells. Cancer Lett 2002; 177:95–100. 125 Littlepage LE, Sternlicht MD, Rougier N, Phillips J, Gallo E, Yu Y et al. Matrix metalloproteinases contribute distinct roles in neuroendocrine prostate carcinogenesis, metastasis, and angiogenesis progression. Cancer Res 2010;70:2224–34. 126 Pozzi A, LeVine WF, Gardner HA. Low plasma levels of matrix metalloproteinase 9 permit increased tumor angiogenesis. Oncogene 2002;21:272–81. 127 Fang C, Wen G, Zhang L, Lin L, Moore A, Wu S et al. An important role of matrix metalloproteinase-8 in angiogenesis in vitro and in vivo. Cardiovasc Res 2013;99:146–55. 128 Bouvet C, Gilbert LA, Girardot D, deBlois D, Moreau P. Different involvement of extracellular matrix components in small and large arteries during chronic NO synthase inhibition. Hypertension 2005;45:432–7. 129 Prado AF, Castro MM, Gerlach RF. Matrix metalloproteinases and hypertension. In: Dhalla NS, Chakraborti S (eds). Role of Protease in Cellular Dysfunction. Spriger, New York, 2014. pg. 279–93. 130 Castro MM, Rizzi E, Figueiredo-Lopes L, Fernandes K, Bendhack LM, Pitol DL et al. Metalloproteinase inhibition ameliorates hypertension and prevents vascular dysfunction and remodeling in renovascular hypertensive rats. Atherosclerosis 2008;198:320–31. 131 Castro MM, Rizzi E, Rodrigues GJ, Ceron CS, Bendhack LM, Gerlach RF et al. Antioxidant treatment reduces matrix metalloproteinase-2-induced vascular changes in renovascular hypertension. Free Radical Biol Med 2009;46:1298–307. 132 Cau SB, Guimaraes DA, Rizzi E, Ceron CS, Souza LL, Tirapelli CR et al. Pyrrolidine dithiocarbamate down-regulates vascular matrix metalloproteinases and ameliorates vascular dysfunction and remodelling in renovascular hypertension. Br J Pharmacol 2011;164:372–81. 133 Ceron CS, Castro MM, Rizzi E, Montenegro MF, Fontana V, Salgado MC et al. Spironolactone and hydrochlorothiazide exert antioxidant effects and reduce vascular matrix metalloproteinase2 activity and expression in a model of renovascular hypertension. Br J Pharmacol 2010;160:77–87. 134 Ceron CS, Rizzi E, Guimaraes DA, Martins-Oliveira A, Cau SB, Ramos J et al. Time course involvement of matrix metalloproteinases in the vascular alterations of renovascular hypertension. Matrix Biol 2012;31:261–70. 135 Guimaraes DA, Rizzi E, Ceron CS, Oliveira AM, Oliveira DM, Castro MM et al. Doxycycline dose-dependently inhibits MMP2-mediated vascular changes in 2K1C hypertension. Basic Clin Pharmacol Toxicol 2011 May;108(5):318–25. doi: 10.1111/j. 1742-7843.2010.00656.x. 136 Marcal DM, Rizzi E, Martins-Oliveira A, Ceron CS, Guimaraes DA, Gerlach RF et al. Comparative study on antioxidant effects and vascular matrix metalloproteinase-2 downregulation by dihydropyridines in renovascular hypertension. Naunyn Schmiedebergs Arch Pharmacol. 2011;383:35–44. 137 Martinez ML, Castro MM, Rizzi E, Fernandes K, Demacq C, Bendhack LM et al. Lercanidipine reduces matrix metalloproteinase-2 activity and reverses vascular dysfunction in renovascular hypertensive rats. Eur J Pharmacol 2008;591:224–30. 138 Martins-Oliveira A, Castro MM, Oliveira DM, Rizzi E, Ceron CS, Guimaraes D et al. Contrasting effects of aliskiren versus losartan on hypertensive vascular remodeling. Int J Cardiol 2012;167(4):1199–205. doi: 10.1016/j.ijcard.2012.03.137.

MiniReview

139 Rizzi E, Castro MM, Ceron CS, Neto-Neves EM, Prado CM, Rossi MA et al. Tempol inhibits TGF-beta and MMPs upregulation and prevents cardiac hypertensive changes. Int J Cardiol 2011;165(1):165–73. 140 Rizzi E, Castro MM, Prado CM, Silva CA, Fazan R Jr, Rossi MA et al. Matrix metalloproteinase inhibition improves cardiac dysfunction and remodeling in 2-kidney, 1-clip hypertension. J Card Fail 2010;16:599–608. 141 Ceron CS, Rizzi E, Guimaraes DA, Martins-Oliveira A, Gerlach RF, Tanus-Santos JE. Nebivolol attenuates prooxidant and profibrotic mechanisms involving TGF-beta and MMPs, and decreases vascular remodeling in renovascular hypertension. Free Radical Biol Med 2013;65:47–56. 142 Fontana V, Silva PS, Izidoro-Toledo TC, Biagi C, Oliveira EB, Gerlach RF et al. Comprehensive evaluation of the effects of enalapril on matrix metalloproteinases levels in hypertension. Cardiovasc Drugs Ther 2012;26:511–9. 143 Yamamoto D, Takai S, Hirahara I, Kusano E. Captopril directly inhibits matrix metalloproteinase-2 activity in continuous ambulatory peritoneal dialysis therapy. Clin Chim Acta 2010;411:762–4. 144 Sorbi D, Fadly M, Hicks R, Alexander S, Arbeit L. Captopril inhibits the 72 kDa and 92 kDa matrix metalloproteinases. Kidney Int 1993;44:1266–72. 145 Reinhardt D, Sigusch HH, Hensse J, Tyagi SC, Korfer R, Figulla HR. Cardiac remodelling in end stage heart failure: upregulation of matrix metalloproteinase (MMP) irrespective of the underlying disease, and evidence for a direct inhibitory effect of ACE inhibitors on MMP. Heart 2002;88:525–30. 146 Brower GL, Levick SP, Janicki JS. Inhibition of matrix metalloproteinase activity by ACE inhibitors prevents left ventricular remodeling in a rat model of heart failure. Am J Physiol Heart Circ Physiol 2007;292:H3057–64. 147 Kuntze LB, Antonio RC, Izidoro-Toledo TC, Meschiari CA, Tanus-Santos JE, Gerlach RF. Captopril and lisinopril only inhibit matrix metalloproteinase-2 (MMP-2) activity at millimolar concentrations. Basic Clin Pharmacol Toxicol 2014;114:233–9. 148 Carmignani M, Volpe AR, Boscolo P, Qiao N, Di Gioacchino M, Grilli A et al. Catcholamine and nitric oxide systems as targets of chronic lead exposure in inducing selective functional impairment. Life Sci 2000;68:401–15. 149 Tsao DA, Yu HS, Cheng JT, Ho CK, Chang HR. The change of beta-adrenergic system in lead-induced hypertension. Toxicol Appl Pharmacol 2000;164:127–33. 150 Vaziri ND, Ding Y, Ni Z. Compensatory up-regulation of nitricoxide synthase isoforms in lead-induced hypertension; reversal by a superoxide dismutase-mimetic drug. J Pharmacol Exp Ther 2001;298:679–85. 151 Kasperczyk S, Dziwisz M, Kasperczyk A, Birkner E. Influence of lead exposure on arterial hypertension. Wiad Lek 2002;55 (Suppl 1):230–4. 152 Martin D, Glass TA, Bandeen-Roche K, Todd AC, Shi W, Schwartz BS. Association of blood lead and tibia lead with blood pressure and hypertension in a community sample of older adults. Am J Epidemiol 2006;163:467–78. 153 Neuberger JS, Mulhall M, Pomatto MC, Sheverbush J, Hassanein RS. Health problems in Galena, Kansas: a heavy metal mining Superfund site. Sci Total Environ 1990;94:261–72. 154 Rizzi E, Castro MM, Fernandes K, Barbosa F Jr, Arisi GM, Garcia-Cairasco N et al. Evidence of early involvement of matrix metalloproteinase-2 in lead-induced hypertension. Arch Toxicol 2009;83:439–49. 155 Martinez ML, Rizzi E, Castro MM, Fernandes K, Bendhack LM, Gerlach RF et al. Lercanidipine decreases vascular matrix metalloproteinase-2 activity and protects against vascular dysfunction in diabetic rats. Eur J Pharmacol 2008;599:110–6.

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)

MiniReview

MATRIX METALLOPROTEINASES ARE INVOLVED IN CARDIOVASCULAR DISEASES

156 Nagareddy P, Rajput P, Vasudevan H, McClure B, Kumar U, Macleod K et al. Inhibition of matrix metalloproteinase-2 improves endothelial function and prevents hypertension in insulin resistant rats. Br J Pharmacol 2012 Feb;165(3):705–15. doi: 10.1111/j.1476-5381.2011.01583.x. 157 Zeydanli EN, Kandilci HB, Turan B. Doxycycline ameliorates vascular endothelial and contractile dysfunction in the thoracic aorta of diabetic rats. Cardiovasc Toxicol 2011;11:134–47. 158 Castro MM, Rizzi E, Ceron CS, Guimaraes DA, Rodrigues GJ, Bendhack LM et al. Doxycycline ameliorates 2K-1C hypertension-induced vascular dysfunction in rats by attenuating oxidative stress and improving nitric oxide bioavailability. Nitric Oxide 2012;26:162–8. 159 Antonio RC, Ceron CS, Rizzi E, Coelho EB, Tanus-Santos JE, Gerlach RF. Antioxidant effect of doxycycline decreases MMP activity and blood pressure in SHR. Mol Cell Biochem 2014;386:99–105. 160 Bevers LM, Braam B, Post JA, van Zonneveld AJ, Rabelink TJ, Koomans HA et al. Tetrahydrobiopterin, but not L-arginine, decreases NO synthase uncoupling in cells expressing high levels of endothelial NO synthase. Hypertension 2006;47:87–94. 161 Gornik HL, Creager MA. Arginine and endothelial and vascular health. J Nutr 2004;134:2880S–7S; discussion 95S. 162 Silva BR, Pernomian L, Bendhack LM. Contribution of oxidative stress to endothelial dysfunction in hypertension. Front Physiol 2012;3:441. 163 Thomas DD, Ridnour LA, Isenberg JS, Flores-Santana W, Switzer CH, Donzelli S et al. The chemical biology of nitric oxide: implications in cellular signaling. Free Radical Biol Med 2008;45:18–31. 164 Vasquez-Vivar J, Kalyanaraman B, Martasek P, Hogg N, Masters BS, Karoui H et al. Superoxide generation by endothelial nitric oxide synthase: the influence of cofactors. Proc Natl Acad Sci USA 1998;95:9220–5. 165 Camp TM, Smiley LM, Hayden MR, Tyagi SC. Mechanism of matrix accumulation and glomerulosclerosis in spontaneously hypertensive rats. J Hypertens 2003;21:1719–27. 166 DeLano FA, Schmid-Schonbein GW. Proteinase activity and receptor cleavage: mechanism for insulin resistance in the spontaneously hypertensive rat. Hypertension 2008;52:415–23. 167 Fontana V, Silva PS, Belo VA, Antonio RC, Ceron CS, Biagi C et al. Consistent alterations of circulating matrix metalloproteinases levels in untreated hypertensives and in spontaneously hypertensive rats: a relevant pharmacological target. Basic Clin Pharmacol Toxicol 2011;109:130–7. 168 Tran ED, DeLano FA, Schmid-Schonbein GW. Enhanced matrix metalloproteinase activity in the spontaneously hypertensive rat: VEGFR-2 cleavage, endothelial apoptosis, and capillary rarefaction. J Vasc Res 2010;47:423–31. 169 Rodrigues SF, Tran ED, Fortes ZB, Schmid-Schonbein GW. Matrix metalloproteinases cleave the beta2-adrenergic receptor in spontaneously hypertensive rats. Am J Physiol Heart Circ Physiol 2010;299:H25–35. 170 Wu KI, Schmid-Schonbein GW. Nuclear factor kappa B and matrix metalloproteinase induced receptor cleavage in the spontaneously hypertensive rat. Hypertension 2011;57:261–8. 171 Su J, Palen DI, Lucchesi PA, Matrougui K. Mice lacking the gene encoding for MMP-9 and resistance artery reactivity. Biochem Biophys Res Commun 2006;349:1177–81. 172 Hao L, Du M, Lopez-Campistrous A, Fernandez-Patron C. Agonist-induced activation of matrix metalloproteinase-7 promotes vasoconstriction through the epidermal growth factor-receptor pathway. Circ Res 2004;94:68–76. 173 Fernandez-Patron C, Radomski MW, Davidge ST. Vascular matrix metalloproteinase-2 cleaves big endothelin-1 yielding a novel vasoconstrictor. Circ Res 1999;85:906–11.

313

174 Fernandez-Patron C, Stewart KG, Zhang Y, Koivunen E, Radomski MW, Davidge ST. Vascular matrix metalloproteinase-2dependent cleavage of calcitonin gene-related peptide promotes vasoconstriction. Circ Res 2000;87:670–6. 175 Martinez A, Oh HR, Unsworth EJ, Bregonzio C, Saavedra JM, Stetler-Stevenson WG et al. Matrix metalloproteinase-2 cleavage of adrenomedullin produces a vasoconstrictor out of a vasodilator. Biochem J 2004;383:413–8. 176 Fernandez-Patron C, Radomski MW, Davidge ST. Role of matrix metalloproteinase-2 in thrombin-induced vasorelaxation of rat mesenteric arteries. Am J Physiol Heart Circ Physiol 2000;278:H1473–9. 177 Matsusaka H, Ide T, Matsushima S, Ikeuchi M, Kubota T, Sunagawa K et al. Targeted deletion of matrix metalloproteinase 2 ameliorates myocardial remodeling in mice with chronic pressure overload. Hypertension 2006;47:711–7. 178 Rizzi E, Ceron CS, Guimaraes DA, Prado CM, Rossi MA, Gerlach RF et al. Temporal changes in cardiac matrix metalloproteinase activity, oxidative stress, and TGF-beta in renovascular hypertension-induced cardiac hypertrophy. Exp Mol Pathol 2013;94:1–9. 179 Zhou HZ, Ma X, Gray MO, Zhu BQ, Nguyen AP, Baker AJ et al. Transgenic MMP-2 expression induces latent cardiac mitochondrial dysfunction. Biochem Biophys Res Commun 2007;358:189–95. 180 Bergman MR, Teerlink JR, Mahimkar R, Li L, Zhu BQ, Nguyen A et al. Cardiac matrix metalloproteinase-2 expression independently induces marked ventricular remodeling and systolic dysfunction. Am J Physiol Heart Circ Physiol 2007;292: H1847–60. 181 Mahimkar R, Nguyen A, Mann M, Yeh CC, Zhu BQ, Karliner JS et al. Cardiac transgenic matrix metalloproteinase-2 expression induces myxomatous valve degeneration: a potential model of mitral valve prolapse disease. Cardiovasc Pathol 2009;18:253–61. 182 Cheng S, Pollock AS, Mahimkar R, Olson JL, Lovett DH. Matrix metalloproteinase 2 and basement membrane integrity: a unifying mechanism for progressive renal injury. FASEB J 2006;20:1898–900. 183 Demmer RT, Desvarieux M. Periodontal infections and cardiovascular disease: the heart of the matter. J Am Dent Assoc 2006;137(Suppl):14S–20S; quiz 38S. 184 Gendron R, Grenier D, Maheu-Robert L. The oral cavity as a reservoir of bacterial pathogens for focal infections. Microbes Infect 2000;2:897–906. 185 Hoen AG, Rollend LG, Papero MA, Carroll JF, Daniels TJ, Mather TN et al. Effects of tick control by acaricide self-treatment of white-tailed deer on host-seeking tick infection prevalence and entomologic risk for Ixodes scapularis-borne pathogens. Vector Borne Zoonotic Dis 2009;9:431–8. 186 Marcaccini AM, Novaes AB Jr, Meschiari CA, Souza SL, Palioto DB, Sorgi CA et al. Circulating matrix metalloproteinase-8 (MMP-8) and MMP-9 are increased in chronic periodontal disease and decrease after non-surgical periodontal therapy. Clin Chim Acta 2009;409:117–22. 187 Ide M, McPartlin D, Coward PY, Crook M, Lumb P, Wilson RF. Effect of treatment of chronic periodontitis on levels of serum markers of acute-phase inflammatory and vascular responses. J Clin Periodontol 2003;30:334–40. 188 Piconi S, Trabattoni D, Luraghi C, Perilli E, Borelli M, Pacei M et al. Treatment of periodontal disease results in improvements in endothelial dysfunction and reduction of the carotid intimamedia thickness. FASEB J 2009;23:1196–204. 189 Gibson FC III, Hong C, Chou HH, Yumoto H, Chen J, Lien E et al. Innate immune recognition of invasive bacteria accelerates atherosclerosis in apolipoprotein E-deficient mice. Circulation 2004;109:2801–6.

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)

314

ALINE AZEVEDO ET AL.

190 Li L, Messas E, Batista EL Jr, Levine RA, Amar S. Porphyromonas gingivalis infection accelerates the progression of atherosclerosis in a heterozygous apolipoprotein E-deficient murine model. Circulation 2002;105:861–7. 191 Madan M, Bishayi B, Hoge M, Messas E, Amar S. Doxycycline affects diet- and bacteria-associated atherosclerosis in an ApoE heterozygote murine model: cytokine profiling implications. Atherosclerosis 2007;190:62–72. 192 Alvarez B, Ruiz C, Chacon P, Alvarez-Sabin J, Matas M. Serum values of metalloproteinase-2 and metalloproteinase-9 as related to unstable plaque and inflammatory cells in patients with greater than 70% carotid artery stenosis. J Vasc Surg 2004;40:469–75. 193 Blankenberg S, Rupprecht HJ, Poirier O, Bickel C, Smieja M, Hafner G et al. Plasma concentrations and genetic variation of matrix metalloproteinase 9 and prognosis of patients with cardiovascular disease. Circulation 2003;107:1579–85.

MiniReview

194 Ferroni P, Basili S, Martini F, Cardarello CM, Ceci F, Di Franco M et al. Serum metalloproteinase 9 levels in patients with coronary artery disease: a novel marker of inflammation. J Investig Med 2003;51:295–300. 195 Kalela A, Koivu TA, Sisto T, Kanervisto J, Hoyhtya M, Sillanaukee P et al. Serum matrix metalloproteinase-9 concentration in angiographically assessed coronary artery disease. Scand J Clin Lab Invest 2002;62:337–42. 196 Renko J, Kalela A, Jaakkola O, Laine S, Hoyhtya M, Alho H et al. Serum matrix metalloproteinase-9 is elevated in men with a history of myocardial infarction. Scand J Clin Lab Invest 2004;64:255–61. 197 Taurino M, Raffa S, Mastroddi M, Visco V, Rizzo L, Torrisi MR et al. Metalloproteinase expression in carotid plaque and its correlation with plasma levels before and after carotid endarterectomy. Vasc Endovascular Surg 2007;41:516–21.

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)

Matrix metalloproteinases are involved in cardiovascular diseases.

This MiniReview describes the essential biochemical and molecular aspects of matrix metalloproteinases (MMPs) and briefly discusses how they engage in...
267KB Sizes 6 Downloads 3 Views