Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Feb 1.
Published in final edited form as: J Mol Cell Cardiol. 2015 Dec 23;91:134–140. doi: 10.1016/j.yjmcc.2015.12.018

Matrix Metalloproteinases as Input and Output Signals for Post-Myocardial Infarction Remodeling

Merry L Lindsey 1,2, Rugmani Padmanabha Iyer 1, Mira Jung 1, Kristine Y DeLeon-Pennell 1, Yonggang Ma 1
PMCID: PMC4764435  NIHMSID: NIHMS750735  PMID: 26721597

Abstract

Despite current optimal therapeutic regimens, approximately one in four patients diagnosed with myocardial infarction (MI) will go on to develop congestive heart failure, and heart failure has a high five-year mortality rate of 50%. Elucidating mechanisms whereby heart failure develops post-MI, therefore, is highly needed. Matrix metalloproteinases (MMPs) are key enzymes involved in post-MI remodeling of the left ventricle (LV). While MMPs process cytokine and extracellular matrix (ECM) substrates to regulate the inflammatory and fibrotic components of the wound healing response to MI, MMPs also serve as upstream signaling initiators with direct actions on cell signaling cascades. In this review, we summarize the current literature regarding MMP roles in post-MI LV remodeling. We also identify the current knowledge gaps and provide templates for experiments to fill these gaps. A more complete understanding of MMP roles, particularly with regards to upstream signaling roles, may provide new strategies to limit adverse LV remodeling.

Keywords: MMP, signaling, extracellular matrix, systems biology, proteomics, extracellular matridomics

Introduction

According to the European Cardiovascular Disease Statistics (www.ehnheart.org/cvd-statistics.html), each year cardiovascular disease (CVD) causes over 4 million deaths in Europe. In the United States, approximately 6 million Americans currently suffer from heart failure, and the high 5-year mortality rate of 50% results in an annual health care cost of >$34 billion.[1] In 70% of heart failure cases, myocardial infarction (MI) is the underlying etiology.[2-9] While immediate reperfusion of the occluded artery is an optimal therapeutic strategy for ST segment elevation MI, the number of MI patients that are not reperfused (due to late presentation or other exclusion criteria) accounts for approximately 250,000 new patients with permanently occluded arteries each year in the United States.[1] Therefore, adverse remodeling of the left ventricle (LV) following MI remains a significant cause of congestive heart failure.

Matrix metalloproteinases (MMPs) are a family of 25 proteolytic enzymes that regulate extracellular matrix (ECM) turnover and inflammatory signaling. While about half of the MMPs have been measured in the post-MI LV and several MMPs (e.g., MMP-2, -7, -9, -12, -14, and -28) have been studied in detail, there remains significant knowledge gaps.[10-19] To provide a framework for this review, we will use the four postulates for cardiac metalloproteinases actions (CarMA) previously developed to define the iterative process for proving MMP causality in post-MI LV remodeling.[20] The four postulates are: 1) the MMP increases post-MI; 2) the MMP stimulates cell signaling in vitro; 3) modulating MMP levels alters LV remodeling; and 4) MMP proteolytic products recapitulate at least partial components of MMP functions. Note that the four postulates match the order and substance of the Koch’s postulates to establish a cause and effect relationship between a microbe and a disease, rather than by how much evidence is currently available to support the postulate. Postulates 1 and 3 have the most literature available to date.

Below, we list the postulate, followed by the published evidence to support it, followed by the critical knowledge gaps that need to be addressed to provide mechanistic insight into MMP roles in the post-MI LV. We give examples of studies performed in mice with genetic deletions of MMP-7, MMP-9, MMP-14, or MMP-28 or inhibitors against MMP-12. We also discuss a few misconceptions that have arisen along the past 20 years of cardiac MMP research. We end with discussion of how the identified knowledge gaps can be filled, providing example experimental design templates including considerations for controls and potential limitations. While each of the four postulates has been shown for about half of the MMPs, there remains a lack of complete understanding for all MMPs. In addition, multiple MMPs have not been evaluated for even the first postulate. While we discuss each of the postulates to provide a comprehensive review, the second postulate (that MMPs stimulate cell signaling directly) provides an exciting new avenue of research that is only now coming to light.

CarMA Postulate 1: The MMP increases post-MI

This is the smoking gun postulate; the first evidence that must be shown is that the MMP is in a location and at a concentration high enough to have an effect. We and others have shown that MMP-2, -7, -8, -9, -12, and -14 increase post-MI.[12, 21-33] In particular, MMP-9 and MMP-12 have been shown to increase at both the gene and protein level, and both pro- and active protein forms are elevated post-MI.[12, 34] Both increase in the infarct at day 1 post-MI and remain elevated through days 5-7 post-MI.[12, 34]

In addition to measuring total amounts of mRNA or protein expression, it is also important to measure quality and location. With the exceptions of MMP-11, MMP-14, and MMP-28, most MMPs are secreted as pro-enzymes that need to be activated for substrate cleavage.[35, 36] Understanding if the MMP observed is in a pro, active, or inactivated/inhibited state provides knowledge useful for assessing function. Monitoring location and cell source are also important components. For example, total MMP-28 concentration actually decreases post-MI, due to the significant loss of cardiomyocytes that are the major pre-MI source of MMP-28 in the myocardium.[37] At the same time, the amount of MMP-28 contributed by the macrophage increases with the infiltration of macrophages into infarct. This postulate, therefore, is fulfilled for macrophage-derived MMP-28.

Originally, MMPs were classified based on the cell type where the MMP was first observed or the substrates first used to evaluate activity, and this led to confusion.[20] For example, MMP-1 was first named collagenase, but this MMP also processes tenascin and aggrecan into fragments.[20] MMP-8 was first termed neutrophil collagenase, whereas subsequent studies have shown macrophages to be robust expressors of this MMP and additional MMP-8 substrates include aggrecan, fibrinogen, CXCL5, and CXCL6.[38-40] MMP-12 was initially termed macrophage metalloelastase, whereas this MMP also processes osteopontin, and MMP-9 has greater enzyme affinity for elastin cleavage than MMP-12.[16, 41-43] In our own studies, we were surprised to find that post-MI LV neutrophils robustly express MMP-12 at day 1 post-MI.[12] Along with MMPs, the tissue inhibitors of metalloproteinases (TIMPs) have also been misunderstood. While TIMP-4 has been termed the cardiac-specifc TIMP due to its high expression in the myocardium, TIMP-4 is also expressed in the kidney, placenta, colon, and testes.[44]

While CarMA postulate 1 is fulfilled for a subset of MMPs, the current knowledge gap 1 is that each MMP has not been evaluated (e.g., MMP-15 and MMP-16) and not all MMPs (e.g., MMP-7 and MMP-28) have been mapped as extensively as others (e.g., MMP-2 and MMP-9).

CarMA Postulate 2: MMP stimulates cell signaling in vitro

This postulate dictates that MI-relevant cells stimulated with an MMP will display biological functions similar to what is observed during LV remodeling in vivo, if that MMP has a causal role. Basically, this postulate raises the possibility that MMPs can serve in direct signaling capacities, which separates this role from that of an enzyme. While there are examples to provide evidence for this postulate, this idea has not been examined in detail.

In vitro, MMP-9 directly activates macrophage polarization to an M1/M2 transition state.[45] MMP-9 stimulation increased the expression of the pro-inflammatory M1 gene Ccl5, but also decreased the expression of M1 markers Ccl3, interleukin (IL)-1β, and IL-6. Transforming growth factor (TGF)β1, an anti-inflammatory M2 marker, was also down-regulated after MMP-9 treatment. MMP-12 stimulates neutrophils in vitro to induce expression of the apoptosis markers, CD44, caspase 3, and caspase 8,[12] and MMP-9 also stimulates capase 3 expression in neutrophils.[46] CD44 regulates apoptosis by interacting with hyaluronic acid and is a critical mechanism in wound healing to clear inflammatory cells from injury sites.[47, 48] MMP-12 can also process CD44 to generate a 15 kDa fragment, indicating a feedback loop. CD44 cleavage prevents the clearance of the CD44 ligand hyaluronic acid, which is a stimulus for inflammation resolution during wound healing.[12] Combined, these results indicate that MMPs can be used as direct stimulating factors as well as output factors. This is an entirely new concept in MMP biology, and future studies evaluating how MMPs activate cell signaling (e.g., direct binding of receptors such as integrins or indirect effects through processing of substrates) are warranted.

In vivo, MMP-7 has been shown to have direct activity on myocardial electrical activity by cleaving connexin-43 in the C-terminal domain.[49] Infusion of recombinant MMP-7 into the jugular vein of mice induced heart block within 60 min of infusion, concomitant with reduced connexin-43 staining at the myocyte to myocyte borders. This study provides direct evidence that connexin-43 is an in vivo substrate of MMP-7 and that its processing results in a pathophysiological phenotype.

Knowledge Gap 2 is that the MMP signaling pathways that regulate cell function have not been mapped. There is a need to identify MMP signaling pathways that regulate post-MI relevant cell functions, including myocyte apoptosis; neutrophil apoptosis and degranulation; macrophage polarization and phagocytosis; and fibroblast proliferation, differentiation, and ECM expression. Included in this knowledge gap is the need to know which receptors are engaged by MMPs and whether the effects are actually directly occuring through receptor engagement and signaling or due to an indirect effect that has not been elucidated (e.g., substrate fragment binding to a receptor or shedding of an inhibitor in the signaling pathway). While this postulate has the largest unknown component, it also is one of the more exciting postulates due to its novelty.

CarMA Postulate 3: Modulation of an MMP alters LV remodeling

This postulate identifies tissue level functions controlled by MMP regulation of individual ECM components or of cell changes. Concentrations of a few specific MMPs (e.g., MMP-9) directly correlate to the extent of LV dysfunction post-MI. The assumption has been that an increase in an MMP is always detrimental and that MMPs should be inhibited, but this has not always been the case as we have recently seen for MMP-12 inhibition.[12] Inhibiting MMP-12 beginning at 3 hours post-MI exacerbates LV dilation and dysfunction, suggesting beneficial components of MMP-12 activity.[12] MMP-12 stimulates in vivo neutrophil apoptosis, as MMP-12 inhibition increased full-length caspase 3 by 58% and reduced cleaved caspase 3 by 50%.[12] CD18 (β2 integrin; ITGB2), a cell adhesion molecule that suppresses neutrophil apoptosis during endothelial transmigration, was elevated at day 7 post-MI with MMP-12 inhibition. This indicates reduced apoptosis and prolonged neutrophil accumulation when this MMP was inhibited.

The combined results from MMP-9 null mice and mice with transgenic overexpression of MMP-9 only in macrophages reveals that both situations paradoxically improve LV remodeling. [24, 50-53] These findings are due to MMP-9 exerting both negative and positive actions in the post-MI LV. MMP-9 deletion attenuates collagen and fibronectin cleavage to attenuate LV dilation; while MMP-9 overexpression promotes processing of angiogenic factors (e.g., plasminogen and collagen IV) to stimulate neovascularization.[34, 54]

MMP-28 deletion was also detrimental to LV remodeling. During aging, LV inflammation increases with age, and MMP-28 deletion further elevates inflammation and extracellular matrix responses, without altering macrophage numbers or collagen content.[55] MMP-28 deletion increased plasma macrophage inflammatory protein (MIP)-1α, MIP-1β and MMP-9 protein concentrations and elevated MIP-1a and MMP-9 gene and protein levels in the LV, indicating a higher overall inflammatory status when this MMP was genetically modified. After MI, MMP-28 deletion aggravated MI-induced LV dysfunction and rupture as a result of both a defective inflammatory response and reduced scar formation.[23] In both aging and MI, MMP-28 deletion suppressed M2 macrophage activation. This leads to Knowledge Gap 3: MMP signaling pathways that regulate the global LV response (structure and function) have not been identified. We need to understand how the compilation of different signals regulated by MMPs are coordinated to regulate LV remodeling.

CarMA Postulate 4: MMP proteolytic products regulate LV remodeling

According to this postulate, MMP cleavage products should directly serve as MMP substitutes in regulating particular aspects of the post-MI LV remodeling phenotype, such that adding back substrate in an MMP null background should recapitulate the MMP wild type phenotype or at least a component of the phenotype. While a number of MMP substrates have been identified and general consensus sequences have been compiled (Table 1), the complete MMP interactome has not been catalogued for any individual MMP. For MMP-2, the current substrate list is expansive and includes hundreds of proteins, while for MMP-28, the current substrate list only includes casein, Nogo-A (a myelin component), and neural cell adhesion molecule-1.[56, 57] Table 2 lists a sampling of MI-relevant substrates proteolytically processed by matrix metalloproteinases (MMPs).[20, 22, 46, 58-64] Below, we discuss a few interesting substrates for MMP-7, MMP-9, and MMP-12.

Table 1. MMP Substrate Consensus Sequences, based on analysis of known cleavage sites. Letters are abbreviations for amino acids. For details, see http://merops.sanger.ac.uk. – is any amino acid.

1 2 3 4 Cleavage
site
5 6 7 8 Number of known
sites examined
MMP-2 - P - - L - - - 3417
MMP-3 - PA A - L - - - 180
MMP-7 - PA - - L - - - 196
MMP-8 G PAS - G L - G - 114
MMP-9 G PA - G L - G - 369
MMP-
12
G PAG AG GA L - GA - 218
MMP-
14
- P - - L - - - 132

Table 2.

Myocardial infarction (Ml)-relevant substrates proteolytically processed by matrix metalloproteinases (MMPs).[20, 22, 46, 58-64]

Substrate MMPs
Aggrecan -2,-9,-3, -8, -14
CD36 -9
CD44 -14
Citrate synthase -9
Collagen -1,-2,-8,-9,-13,-14
Complement C1q -1, -2, -3, -9
Connexin 43 -7
Desmin -9
Endothelin-1 -9
Fibroblast growth factor receptor 1 -2
Fibronectin -2,-7,-9
Fibrinogen -2, -3, -7, -9, -14
Galectin-3 -9, -2
Interleukin-1β -9
Interleukin-8 -9, -1, -2, -3,
Insulin-like growth factor binding protein-3 -1, -2,-3
laminin -9, -2, -3, -14
Latent TGF-β1, -β2, -β3 -2, -9
Monocyte chemotactic proteins-1, -2, -4 -1, -3
Monocyte chemotactic protein-3 -2, -14
Myosin Light Chain 1 -2
Osteoponin (cite Lis) -2, -3, -7, -9
Pro-MMP-2, -9, -13 -9
Platelet factor 4 -9
serpina 1d -9
Tenascin-C -7,-9
Thrombospondin-1 -9
Troponin-I -2
Tumor necrosis factor-α - -1, -2, -3, -7, -9
VEGF -9
Vitronectin -2, -3,-9

In addition to connexin 43, MMP-7 substrates include denatured collagen and fibronectin.[49, 65] Several MMPs have the ability to activate other MMPs. For example, MMP-3, which can activate MMPs -1, -7, and -9 and is considered an early upstream MMP in the cascade.[66] Likewise, MMP-7 can activate in vitro MMPs -1, -2, -8, and -9, but not MMP-13.[67] In MMP-7 null LV, MMP-8 increases and MMP-13 decreases as a compensatory mechanism.[67] The MMP profile switch reveals that MMP-8 accumulation in the absence of MMP-7 downregulates MMP-13 in order to maintain a baseline collagenolytic function and preserve LV wall thickness and dimensions. This interplay between MMP-8 and MMP-13 indicates that these MMPs play reciprocal roles and demonstrates that when designing selective MMP inhibitors, we need to take into consideration potential compensation mechanisms that will regulate parallel MMP types. This also highlights that baseline MMP null phentoypes are seldom unchanged and measuring global phenotypes may not reveal subtle but important differences.

In addition to ECM molecules, MMP-9 cleaves a variety of inflammatory mediators to coordinate their in vivo functions. MMP-9-mediated proteolysis of cytokines and chemokines is one way by which MMP-9 influences leukocyte trafficking and creates positive or negative feedback loops. For example, the N-terminal cleavage of the CXC chemokine IL-8 by MMP-9 increases its chemotactic activity by 10-fold.[68] IL-8 promotes neutrophil chemotaxis which stimulates additional release of MMP-9 demonstrating a positive feedback loop. Similar to MMP-2, MMP-9 also processes a number of intracellular substrates, including citrate synthase.[59] In wild type mice, citrate synthase dramatically decreases by 1 day post-MI due to proteolytic processing by MMP-9, as MMP-9 deletion preserved citrate synthase concentrations in the mitochondria. This assigns a role for MMP-9 in impairing cardiac metabolism in the setting of ischemia.

MMP-12 cleaves immunoglobulins, which are key stimulators of neutrophil degranulation.[69-71] This indicates immunoglobulin processing may be a critical MMP-12 dependent trigger substrate to regulate neutrophil biology. MMP-12, therefore, regulates many aspects of neutrophil biology, including degranulation and apoptosis. While substrate identification is a straightforward process, making the connection between substrate processing and an effect on cell or tissue physiology in the setting of post-MI LV remodeling has only been comprehensively evaluated for MMP-9 generated collagen matricryptin.[61] A Collagen Iα1 matricryptin generated by MMP-9 cleavage at amino acids 1158 and 1159 (p1158/59) reduced LV dilation and facilitates LV remodeling post-MI by regulating scar formation through targeted ECM generation and stimulation of angiogenesis. From this information, we derive Knowledge Gap 4: MMP proteolytic products that regulate LV remodeling have not been catalogued. We need to understand the mechanisms whereby MMP proteolytic products regulate LV remodeling.

How to fill the knowledge gaps

Above, we summarized the current literature base and captured the critical knowledge gaps that remain in our understanding of how MMPs regulate LV remodeling post-MI. Here, we will provide example experimental design templates that would allow these gaps to be filled. We also discuss new technology that is required to help propel this research.

Knowledge gap 1

each MMP has not been evaluated (e.g., MMP-15 and MMP-16) and not all MMPs (e.g., MMP-7 and MMP-28) have been mapped as extensively as others (e.g., MMP-2 and MMP-9). Filling this knowledge gap is technically uncomplicated, as quantative PCR can be used to assess gene levels and there are many commercially available antibodies for all of the MMPs, which allows immunoblotting assessment. While there are no technical challenges, time and location of evaluation need to be considered in the experimental design. Assessing both the remote non-infarcted myocardium and the infarct region during a full time continuum with adequate sample size in each group to have sufficient sample size to detect differences makes these experiments complex. While evaluating at the mRNA level is quite feasible, running this number of samples by immunoblotting is difficult, particularly given issues with intra-blot comparisons. One strategy is to evaluate the time course with a pooled sample strategy, such that each time point is run as one sample, taking into account the caveat that a potential confounder to this strategy is the presence of any outliers in the pooled samples that could mask true effects. From there, one time point can be selected to compare against day 0 naïve or sham controls. What we need to accelerate the closure of this knowledge gap is an MMP protein array that allows quantitation of both pro and active MMP forms and includes the tissue inhibitors of metalloproteinsaes (TIMPs) in a more rapid and accurate way. While there are arrays available for a handful of MMPs, ideally an array that differentiates the pro and active forms of all MMPs and includes all TIMPs would aid in closing this knowledge gap.

Knowledge Gap 2

The MMP signaling pathways that regulate cell function have not been mapped. Table 3 provides an example experimental template for examining direct MMP effects on cell functions. We used MMP stimulation of neutrophil apoptosis and macrophage polarization as examples here.

Table 3.

Experiments to fill knowledge gap 2.

General design:
  1. isolate and culture cells of interest

  2. perform time and dose responses with MMP as stimulus

  3. examine cell response

  4. compare ex vivo and in vivo responses

Cell Example responses
(could be ↑ or ↓)
What to measure
All
  • Global gene expression

  • Global protein expression
    • cellular or secreted
  • Genomics

  • Proteomics

Neutrophil
  • Degranulation

  • Apoptosis

  • Secreted markers

  • Caspases

  • CD44

Macrophage
  • Polarization

  • Phagocytosis

  • Pro- and anti-inflammatory markers

  • CD36; uptake of fluorescent bioparticles1

Fibroblast
  • Proliferation

  • Migration

  • Wound healing

  • ECM production

  • BrdU uptake

  • Transwell mobilization

  • Scrape or heat injury2

  • ECM genes and proteins3

BrdU- 5-Bromo-2′-deoxyuridine; ECM- extracellular matrix

1

e.g., fluorescein-labeled Escherichia coli K-12 bioparticles (Molecular Probes)

2

e.g., Electric Cell-substrate Impedance Sensing System (Applied Biophysics)

3

MI relevant ECM includes collagen, fibronectin, laminin, and matricellular proteins

Neutrophils isolated from the blood or bone marrow and stimulated with an MMP in vitro can be compared to unstimulated cells as a negative control. The cells can be examined for marker expression or functional responses by flow cytometry, qRT-PCR, cytochemistry or proteomic evaluations. Conditioned media can also be examined by ELISA or proteomics, as long as the media does not contain a large concentration of serum (<0.1% is ideal). One positive control would be neutrophils isolated from the infarct region and immediately evaluated.

Macrophages isolated from the peritoneum or monocytes isolated from the blood or bone marrow and stimulated with an MMP in vitro can be compared to unstimulated cells as a negative control. The cells can be examined for M1 and M2 marker expression. An advantage of peritoneal macrophages is that enough cells can be isolated from one mouse to examine 6-8 different conditions, whereas blood monocytes from 6 mice need to be pooled to have the same numbers of isolated cells. The advantage of blood monocytes is that these cells will infiltrate into the LV post-MI, whereas peritoneal macrophages may not fully recapitulate the MI macrophage phenotype.[45, 72] Cells stimulated with interleukin (IL)-1β or lipopolysaccharide (LPS) + interferon (IFN)-γ serve as positive controls for M1 polarization, while IL-4 or IL-10 serve as positive controls for M2 polarization. Other positive controls are macrophages isolated from the infarct region or bone marrow derived macrophages, to compare back how well the in vitro setting recapitulates the in vivo situation. A co-stimulation or sequential stimulation experiment may be warranted, to assess cells under a condition that more closely reflects the in vivo post-MI environment.

Once it is established that the MMP activates (or inactivates) a cell function, the next step is to elucidate the mechanism whereby this occurs. The first consideration is whether this is a substrate mediated enzymatic event, such as MMP processing a ligand or receptor directly. If this is not the case, examining whether the MMP can directly bind to a receptor can be tested.

Target receptors can be assessed by proteomic methods, which provide unbiased appraisal. Using a solid phase extraction of glycopeptides (SPEG) with hydrazide chemistry protocol can be performed to identify glycopeptides in WT and MMP null infarct homogenates or cell culture conditioned media.[73, 74] Since most receptors are glycoproteins, this approach eliminates the potential issue of high levels of albumin in the conditioned media, since albumin is not a glycoprotein and will not be isolated or labeled by this approach. This approach is also useful for ECM-focused analysis (matridomics).[75-78]

Targeted assesssment of candidate receptors is also possible, and surface plasmon resonance can be used to test the in vitro binding kinetics. When testing viability of receptor targets, it is best to first use bioinformatics and simulations to rank candidate evaluation. Comparing homology of MMPs with known receptor stimulators will minimize the number of receptors that need to be tested. Binding simulations would also elucidate receptor candidates that could then be tested in vitro and in vivo.

Knowledge Gap 3

MMP signaling pathways that regulate the global LV response (structure and function) have not been identified. Filling this gap will identify tissue-specific actions, including effects on tissue mechanics.[79] To address this gap, permanent occlusion or reperfusion models are useful, and both have strengths and limitations. In a 2012 Scientific Statement on animal models of heart failure from the American Heart Association, reperfusion and permanent coronary artery ligation were deemed clinically relevant and significant models.[55] While MMP deletion or transgenic stable or conditional mouse models have been evaluated, a translational approach is to infuse or inhibit the MMP in a setting that recapitulates the clinic. A constant infusion protocol can be used to evaluate in vivo responses, and one protocol is to begin at 3 hours post-MI, which will capture the entirety of the inflammatory and wound healing responses. Starting too early post-MI (or pre-MI) changes the design in a way that limits clinical translation. To rule out the possibility that the MMP is regulating an early response (e.g., myocytes or neutrophils) to indirectly affect a later response (e.g., macrophages or fibroblasts), constant infusion beginning at day 2 post-MI can be used to offset this limitation.

Table 4 shows useful variables to assess the LV remodeling phenotype. LV geometry, strain analysis, and function can all be evaluated in vivo by ultrasound, magnetic resonance imaging, or hemodynamic evaluation. The LV (infarct and remote regions) can be frozen for biochemical analysis (gene or protein) or fixed for pathological examination of inflammation, ECM turnover, and scar formation. As a control for the MMP infusion, lungs and right ventricle can be weighed and myocyte cross sectional areas measured to rule out if effects are due to pulmonary issues and also assess heart hypertrophy and failure status. Consideration should also be given from the dose used; the dose should provide over-expression without being supra-pathological.

Table 4.

Measuring the LV remodeling phenotype

Phenotype Variables measured
Infarct structure; LV function;
hemodynamics; cardiac
metabolism
LV geometry, strain analysis, and ejection fraction by
echocardiography, magnetic resonance imaging, or
hemodynamic assessment
Hypertrophy and heart failure Organ weights at necropsy- heart and lungs; myocyte cross
sectional areas
Infarct area Viable and infarct myocardium areas by metabolic staining;
hematoxylin and eosin stained histological sections
Scar formation Picrosirius red stained histological sections
Inflammation and its
resolution
Pro- and anti-inflammatory mediators (gene and protein)
Neutrophils neutrophil extracellular traps; tissue destruction;
degranulation markers
Macrophage polarization M1 and M2 markers (gene and protein)
Fibroblast activation α-smooth muscle actin levels; extracellular matrix
production and turnover (MMP concentrations and ECM
fragment formation)
Neovascularization Griffonia (Bandeiraea) Simplicifolia Lectin 1 staining of
histological sections; angiogenesis markers (e.g., VEGF)
Cell-cell interactions Co-culture; secretome composition by mass spectrometry

Knowledge Gap 4

MMP proteolytic products that regulate LV remodeling have not been catalogued. Evaluating if substrate proteolyic products regulate the same signaling pathways will show direct cause and effect relationships. An experimental flow chart for how to answer this question is shown in Figure 1. We used this flow chart recently to examine the effects of a novel collagen fragment in post-MI LV remodeling.[61]

Figure 1.

Figure 1

Once a candidate MMP substrate is identified, cleavage can be confirmed using an in vitro assay consisting of the MMP and a recombinant of the candidate protein. Using mass spectrometry, the cleavage site(s) can be mapped. Once sites are known, comparing the literature for similar matricryptins will provide insights into putative functions. For example, fibronectin and laminin matricryptins have been studied extensively in the angiogenesis field.[83] To test for biological function, peptides 15-20 amino acids in length that are immediately upstream or downstream of the cleavage site can be commercially generated. If many cleavage sites exist, computational analysis can help to focus on the most likely candidates. Starting with in vitro cell-based assays is higher throughput and more cost effective and will eliminate candidates that have no activity on any cell type of interest (i.e., cardiomyocytes, leukocytes, fibroblasts, or endothelial cells). Using small peptides to test biological activities of naturally occurring fragments bypasses difficulties associated with larger peptide synthesis. A short peptide is easier to synthesize, purify, and concentrate, and a small size peptide limits response variability, which can occur with a longer peptide that may possess multiple activities. Using a small peptide increases the probability that an effect is a direct response to the sequence and not due to peptide structural properties.[92]

Future Directions and Conclusions

While this review focused on MMPs in the post-MI setting, a similar template can be applied for other relevant molecular components and as a broad application to other diseases that involve MMPs. For example, the A Disintegrin and Metalloproteinase (ADAM) family of enzymes are important for post-MI healing, and this family has been reviewed by others, including the Kassiri lab.[80, 81] Because a substantial amount of work remains to be done to establish cause and effect relationships between MMPs and LV remodeling components, we focused on MMP effects on single cell actions. Gene deletion or transgene strategies have been used to decrease or increase MMP expression, but these approaches occur in an artificial setting that is not directly translational. While it is relatively simple to overexpress MMPs systemically with recombinant proteins or peptides, tools available to limit MMP function are limited and suffer from low selectivity. Table 5 lists a sampling of known MMP inhibitors. Of note, issues with inhibitor selectivity and specificity continue; often inhibitors that are relatively specific at lower doses become broad spectrum inhibitors at higher doses.

Table 5.

Matrix metalloproteinase (MMP) inhibitors- note that very few are selective for specific MMPs.[88-91]

Inhibitor MMPs inhibited
CGS27023A MMP-2, MMP-3, MMP-9, MMP-12
matlystatin A MMP-2, MMP-9
matlystatin B MMP-2, MMP-9
ONO-4817 MMP-2, MMP-3, MMP-7, MMP-8, MMP-9,
MMP-12
Ro 28-2653 MMP-2, MMP-9, MMP-14
SB-3CT MMP-2, MMP-9
Doxycycline MMP-2, MMP-9
Galardin MMP-3, MMP-14
α1(I-III)Glyφ{PO2H-CH2}Leu THPI MMP-9
RXP470.1 MMP-12
Imidapril MMP-9
Captopril and Ramiprilate MMP-2 and MMP-9
Lisinopril MMP-9

A future direction is to incorporate intercellular communication to understand how one MMP can activate a cell response that feeds into activating another cell type. Computational models at the cellular scale, such as fibroblast models, will be useful in achieving this goal.[82] For example myocyte-neutrophil, neutrophil-macrophage, and macrophage-fibroblasts are all interconnected in ways that remain to be fully elucidated. The MatrixDB is an extracellular matrix interaction database that will continue to be useful as connections are made among ECM components.[83] In the review by Francis et.al., the Ripplinger lab discusses the interconnections among inflammation, fibrosis, and arrhythmias, highlighting the complexity of the system.[84] Understanding the interconnection between inflammation and fibrosis is an important future avenue of research.[85-87]

In conclusion, understanding the full complement of MMP roles will not only provide us with new therapeutic strategies to treat patients post-MI but will also provide novel diagnostic markers for the early detection of adverse LV remodeling as an early harbinger of ensuing heart failure.

Highlights.

  • MMPs are key enzymes involved in post-MI remodeling of the left ventricle (LV).

  • MMPs process extracellular matrix (ECM) and cytokine substrates.

  • MMPs also serve as upstream initiators with direct actions on cell signaling.

  • We review current MMP knowledge gaps in the cardiac remodeling field.

Acknowledgements

This work was supported by the National Institutes of Health HHSN 268201000036C (N01-HV-00244), HL075360, and GM114833, and the Biomedical Laboratory Research and Development Service of the Veterans Affairs Office of Research and Development Award 5I01B×000505 to MLL; American Heart Association Postdoctoral Grant 14POST18770012 to RPI and Scientist Development Grant 15SDG22930009 to YM; and P01HL051971 and P20GM104357.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • [1].Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, et al. Heart disease and stroke statistics--2015 update: a report from the American Heart Association. Circulation. 2015;131:e29–322. doi: 10.1161/CIR.0000000000000152. [DOI] [PubMed] [Google Scholar]
  • [2].Roger VL, Go AS, Lloyd-Jones DM, Adams RJ, Berry JD, Brown TM, et al. Heart disease and stroke statistics--2011 update: a report from the American Heart Association. Circulation. 2011;123:e18–e209. doi: 10.1161/CIR.0b013e3182009701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Solomon SD, Sutton MSJ, Lamas GA, Plappert T, Rouleau JL, Skali H, et al. Ventricular Remodeling Does Not Accompany the Development of Heart Failure in Diabetic Patients After Myocardial Infarction. Circulation. 2002;106:1251–5. doi: 10.1161/01.cir.0000032313.82552.e3. [DOI] [PubMed] [Google Scholar]
  • [4].Lavine SJ. Prediction of heart failure post myocardial infarction: comparison of ejection fraction, transmitral filling parameters, and the index of myocardial performance. Echocardiography. 2003;20:691–701. doi: 10.1111/j.0742-2822.2003.02156.x. [DOI] [PubMed] [Google Scholar]
  • [5].Hellermann JP, Jacobsen SJ, Redfield MM, Reeder GS, Weston SA, Roger VL. Heart failure after myocardial infarction: clinical presentation and survival. Eur J Heart Fail. 2005;7:119–25. doi: 10.1016/j.ejheart.2004.04.011. [DOI] [PubMed] [Google Scholar]
  • [6].Hellermann JP, Goraya TY, Jacobsen SJ, Weston SA, Reeder GS, Gersh BJ, et al. Incidence of heart failure after myocardial infarction: is it changing over time? Am J Epidemiol. 2003;157:1101–7. doi: 10.1093/aje/kwg078. [DOI] [PubMed] [Google Scholar]
  • [7].Anavekar NS, McMurray JJV, Velazquez EJ, Solomon SD, Kober L, Rouleau J-L, et al. Relation between Renal Dysfunction and Cardiovascular Outcomes after Myocardial Infarction. N Engl J Med. 2004;351:1285–95. doi: 10.1056/NEJMoa041365. [DOI] [PubMed] [Google Scholar]
  • [8].Sutton MSJ, Pfeffer MA, Moye L, Plappert T, Rouleau JL, Lamas G, et al. Cardiovascular Death and Left Ventricular Remodeling Two Years After Myocardial Infarction: Baseline Predictors and Impact of Long-term Use of Captopril: Information From the Survival and Ventricular Enlargement (SAVE) Trial. Circulation. 1997;96:3294–9. doi: 10.1161/01.cir.96.10.3294. [DOI] [PubMed] [Google Scholar]
  • [9].Ezekowitz JA, Kaul P, Bakal JA, Armstrong PW, Welsh RC, McAlister FA. Declining In-Hospital Mortality and Increasing Heart Failure Incidence in Elderly Patients With First Myocardial Infarction. J Am Coll Cardiol. 2009;53:13–20. doi: 10.1016/j.jacc.2008.08.067. [DOI] [PubMed] [Google Scholar]
  • [10].Wells JM, Gaggar A, Blalock JE. MMP generated matrikines. Matrix biology : journal of the International Society for Matrix Biology. 2015 doi: 10.1016/j.matbio.2015.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Lagente V, Le Quement C, Boichot E. Macrophage metalloelastase (MMP-12) as a target for inflammatory respiratory diseases. Expert opinion on therapeutic targets. 2009;13:287–95. doi: 10.1517/14728220902751632. [DOI] [PubMed] [Google Scholar]
  • [12].Iyer RP, Patterson NL, Zouein FA, Ma Y, Dive V, de Castro Bras LE, et al. Early matrix metalloproteinase-12 inhibition worsens post-myocardial infarction cardiac dysfunction by delaying inflammation resolution. International journal of cardiology. 2015;185:198–208. doi: 10.1016/j.ijcard.2015.03.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Marchant DJ, Bellac CL, Moraes TJ, Wadsworth SJ, Dufour A, Butler GS, et al. A new transcriptional role for matrix metalloproteinase-12 in antiviral immunity. Nature medicine. 2014;20:493–502. doi: 10.1038/nm.3508. [DOI] [PubMed] [Google Scholar]
  • [14].Wang X, Liang J, Koike T, Sun H, Ichikawa T, Kitajima S, et al. Overexpression of Human Matrix Metalloproteinase-12 Enhances the Development of Inflammatory Arthritis in Transgenic Rabbits. Am J Pathol. 2004;165:1375–83. doi: 10.1016/S0002-9440(10)63395-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].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 doi: 10.1161/01.CIR.0000121728.14930.DE. 01.CIR.0000121728.14930.DE. [DOI] [PubMed] [Google Scholar]
  • [16].Hou P, Troen T, Ovejero MC, Kirkegaard T, Andersen TL, Byrjalsen I, et al. Matrix metalloproteinase-12 (MMP-12) in osteoclasts: new lesson on the involvement of MMPs in bone resorption. Bone. 2004;34:37–47. doi: 10.1016/j.bone.2003.08.011. [DOI] [PubMed] [Google Scholar]
  • [17].Matsumoto S-i, Kobayashi T, Katoh M, Saito S, Ikeda Y, Kobori M, et al. Expression and Localization of Matrix Metalloproteinase-12 in the Aorta of Cholesterol-Fed Rabbits. Am J Pathology. 1998;153:109–19. doi: 10.1016/s0002-9440(10)65551-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Nenan S, Planquois J-M, Berna P, De Mendez I, Hitier S, Shapiro SD, et al. Analysis of the inflammatory response induced by rhMMP-12 catalytic domain instilled in mouse airways. International Immunopharmacology. 2005;5:511–24. doi: 10.1016/j.intimp.2004.10.011. [DOI] [PubMed] [Google Scholar]
  • [19].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. doi: 10.1016/j.surg.2004.12.004. [DOI] [PubMed] [Google Scholar]
  • [20].Iyer RP, Patterson NL, Fields GB, Lindsey ML. The history of matrix metalloproteinases: milestones, myths, and misperceptions. American journal of physiology: Heart and circulatory physiology. 2012;303:H919–30. doi: 10.1152/ajpheart.00577.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Peterson JT, Li H, Dillon L, Bryant JW. Evolution of matrix metalloprotease and tissue inhibitor expression during heart failure progression in the infarcted rat. Cardiovascular research. 2000;46:307–15. doi: 10.1016/s0008-6363(00)00029-8. [DOI] [PubMed] [Google Scholar]
  • [22].Lindsey ML, Zamilpa R. Temporal and spatial expression of matrix metalloproteinases and tissue inhibitors of metalloproteinases following myocardial infarction. Cardiovasc Ther. 2012;30:31–41. doi: 10.1111/j.1755-5922.2010.00207.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Ma Y, Halade GV, Zhang J, Ramirez TA, Levin D, Voorhees A, et al. Matrix metalloproteinase-28 deletion exacerbates cardiac dysfunction and rupture after myocardial infarction in mice by inhibiting M2 macrophage activation. Circulation research. 2013;112:675–88. doi: 10.1161/CIRCRESAHA.111.300502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Lindsey ML, Escobar GP, Dobrucki LW, Goshorn DK, Bouges S, Mingoia JT, et al. Matrix metalloproteinase-9 gene deletion facilitates angiogenesis after myocardial infarction. American journal of physiology Heart and circulatory physiology. 2006;290:H232–H9. doi: 10.1152/ajpheart.00457.2005. [DOI] [PubMed] [Google Scholar]
  • [25].Zavadzkas JA, Mukherjee R, Rivers WT, Patel RK, Meyer EC, Black LE, et al. Direct regulation of membrane type 1 matrix metalloproteinase following myocardial infarction causes changes in survival, cardiac function, and remodeling. American journal of physiology: Heart and circulatory physiology. 2011;301:H1656–66. doi: 10.1152/ajpheart.00141.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Deschamps AM, Yarbrough WM, Squires CE, Allen RA, McClister DM, Dowdy KB, et al. Trafficking of the membrane type-1 matrix metalloproteinase in ischemia and reperfusion: relation to interstitial membrane type-1 matrix metalloproteinase activity. Circulation. 2005;111:1166–74. doi: 10.1161/01.CIR.0000157149.71297.3A. [DOI] [PubMed] [Google Scholar]
  • [27].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. American journal of physiology: Heart and circulatory physiology. 2006;291:H1838–46. doi: 10.1152/ajpheart.00026.2006. [DOI] [PubMed] [Google Scholar]
  • [28].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. American journal of physiology: Heart and circulatory physiology. 2006;291:H2216–28. doi: 10.1152/ajpheart.01343.2005. [DOI] [PubMed] [Google Scholar]
  • [29].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. American journal of physiology: Heart and circulatory physiology. 2007;292:H1847–60. doi: 10.1152/ajpheart.00434.2006. [DOI] [PubMed] [Google Scholar]
  • [30].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. doi: 10.1371/journal.pone.0034177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Lovett DH, Mahimkar R, Raffai RL, Cape L, Zhu BQ, Jin ZQ, et al. N-terminal truncated intracellular matrix metalloproteinase-2 induces cardiomyocyte hypertrophy, inflammation and systolic heart failure. PloS one. 2013;8:e68154. doi: 10.1371/journal.pone.0068154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Ali MA, Schulz R. Activation of MMP-2 as a key event in oxidative stress injury to the heart. Front Biosci. 2009;14:699–716. doi: 10.2741/3274. [DOI] [PubMed] [Google Scholar]
  • [33].Hughes BG, Fan X, Cho WJ, Schulz R. MMP-2 is localized to the mitochondria-associated membrane of the heart. American journal of physiology: Heart and circulatory physiology. 2014;306:H764–70. doi: 10.1152/ajpheart.00909.2013. [DOI] [PubMed] [Google Scholar]
  • [34].Lindsey ML, Escobar GP, Dobrucki LW, Goshorn DK, Bouges S, Mingoia JT, et al. Matrix metalloproteinase-9 gene deletion facilitates angiogenesis after myocardial infarction. American journal of physiology: Heart and circulatory physiology. 2006;290:H232–9. doi: 10.1152/ajpheart.00457.2005. [DOI] [PubMed] [Google Scholar]
  • [35].Rosenblum G, Meroueh S, Toth M, Fisher JF, Fridman R, Mobashery S, et al. Molecular structures and dynamics of the stepwise activation mechanism of a matrix metalloproteinase zymogen: challenging the cysteine switch dogma. J Am Chem Soc. 2007;129:13566–74. doi: 10.1021/ja073941l. [DOI] [PubMed] [Google Scholar]
  • [36].Rosenblum G, Van den Steen PE, Cohen SR, Bitler A, Brand DD, Opdenakker G, et al. Direct visualization of protease action on collagen triple helical structure. PloS one. 2010;5:e11043. doi: 10.1371/journal.pone.0011043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Ma Y, Halade GV, Zhang J, Ramirez TA, Levin D, Voorhees A, et al. Matrix metalloproteinase-28 deletion exacerbates cardiac dysfunction and rupture after myocardial infarction in mice by inhibiting M2 macrophage activation. Circulation research. 2013;112:675–88. doi: 10.1161/CIRCRESAHA.111.300502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Lindsey ML, Zamilpa R. Temporal and spatial expression of matrix metalloproteinases and tissue inhibitors of metalloproteinases following myocardial infarction. Cardiovascular therapeutics. 2012;30:31–41. doi: 10.1111/j.1755-5922.2010.00207.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Hiller O, Lichte A, Oberpichler A, Kocourek A, Tschesche H. Matrix metalloproteinases collagenase-2, macrophage elastase, collagenase-3, and membrane type 1-matrix metalloproteinase impair clotting by degradation of fibrinogen and factor XII. The Journal of biological chemistry. 2000;275:33008–13. doi: 10.1074/jbc.M001836200. [DOI] [PubMed] [Google Scholar]
  • [40].Van Den Steen PE, Wuyts A, Husson SJ, Proost P, Van Damme J, Opdenakker G. Gelatinase B/MMP-9 and neutrophil collagenase/MMP-8 process the chemokines human GCP-2/CXCL6, ENA-78/CXCL5 and mouse GCP-2/LIX and modulate their physiological activities. European journal of biochemistry / FEBS. 2003;270:3739–49. doi: 10.1046/j.1432-1033.2003.03760.x. [DOI] [PubMed] [Google Scholar]
  • [41].Skjot-Arkil H, Clausen RE, Nguyen QH, Wang Y, Zheng Q, Martinez FJ, et al. Measurement of MMP-9 and -12 degraded elastin (ELM) provides unique information on lung tissue degradation. BMC pulmonary medicine. 2012;12:34. doi: 10.1186/1471-2466-12-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Van Doren SR. Matrix metalloproteinase interactions with collagen and elastin. Matrix biology : journal of the International Society for Matrix Biology. 2015;44-46:224–31. doi: 10.1016/j.matbio.2015.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Lang R, Kocourek A, Braun M, Tschesche H, Huber R, Bode W, et al. Substrate specificity determinants of human macrophage elastase (MMP-12) based on the 1.1 A crystal structure. Journal of molecular biology. 2001;312:731–42. doi: 10.1006/jmbi.2001.4954. [DOI] [PubMed] [Google Scholar]
  • [44].Greene J, Wang M, Liu YE, Raymond LA, Rosen C, Shi YE. Molecular cloning and characterization of human tissue inhibitor of metalloproteinase 4. The Journal of biological chemistry. 1996;271:30375–80. doi: 10.1074/jbc.271.48.30375. [DOI] [PubMed] [Google Scholar]
  • [45].Ma Y, Chiao YA, Clark R, Flynn ER, Yabluchanskiy A, Ghasemi O, et al. Deriving a cardiac ageing signature to reveal MMP-9-dependent inflammatory signalling in senescence. Cardiovascular research. 2015;106:421–31. doi: 10.1093/cvr/cvv128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].DeLeon-Pennell KY, Tian Y, Zhang B, Cates CA, Padmanabhan Iyer R, Cannon P, et al. CD36 Is a Matrix Metalloproteinase-9 Substrate that Stimulates Neutrophil Apoptosis and Removal during Cardiac Remodeling. Circulation Cardiovascular genetics. 2015 doi: 10.1161/CIRCGENETICS.115.001249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Huebener P, Abou-Khamis T, Zymek P, Bujak M, Ying X, Chatila K, et al. CD44 is critically involved in infarct healing by regulating the inflammatory and fibrotic response. J Immunol. 2008;180:2625–33. doi: 10.4049/jimmunol.180.4.2625. [DOI] [PubMed] [Google Scholar]
  • [48].Teder P, Vandivier RW, Jiang D, Liang J, Cohn L, Pure E, et al. Resolution of lung inflammation by CD44. Science. 2002;296:155–8. doi: 10.1126/science.1069659. [DOI] [PubMed] [Google Scholar]
  • [49].Lindsey ML, Escobar GP, Mukherjee R, Goshorn DK, Sheats NJ, Bruce JA, et al. Matrix Metalloproteinase-7 Affects Connexin-43 Levels, Electrical Conduction, and Survival After Myocardial Infarction. Circulation. 2006;113:2919–28. doi: 10.1161/CIRCULATIONAHA.106.612960. [DOI] [PubMed] [Google Scholar]
  • [50].Chiao YA, Ramirez TA, Zamilpa R, Okoronkwo SM, Dai Q, Zhang J, et al. Matrix metalloproteinase-9 deletion attenuates myocardial fibrosis and diastolic dysfunction in ageing mice. Cardiovascular research. 2012;96:444–55. doi: 10.1093/cvr/cvs275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Zamilpa R, Ibarra J, de Castro Bras LE, Ramirez TA, Nguyen N, Halade GV, et al. Transgenic overexpression of matrix metalloproteinase-9 in macrophages attenuates the inflammatory response and improves left ventricular function post-myocardial infarction. Journal of molecular and cellular cardiology. 2012;53:599–608. doi: 10.1016/j.yjmcc.2012.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].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 experimental myocardial infarction. The Journal of clinical investigation. 2000;106:55–62. doi: 10.1172/JCI8768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Zamilpa R, Ibarra J, de Castro Bras LE, Ramirez TA, Nguyen N, Halade GV, et al. Transgenic overexpression of matrix metalloproteinase-9 in macrophages attenuates the inflammatory response and improves left ventricular function post-myocardial infarction. Journal of molecular and cellular cardiology. 2012;53:599–608. doi: 10.1016/j.yjmcc.2012.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Zamilpa R, Lopez EF, Chiao YA, Dai Q, Escobar GP, Hakala K, et al. Proteomic analysis indentifies in vivo candidate matrix metalloproteinase-9 substrates in the left ventricle post-myocardial infarction. Proteomics. 2010;10:2214–23. doi: 10.1002/pmic.200900587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Houser SR, Margulies KB, Murphy AM, Spinale FG, Francis GS, Prabhu SD, et al. Animal Models of Heart Failure: A Scientific Statement From the American Heart Association. Circ Res. 2012;111:131–50. doi: 10.1161/RES.0b013e3182582523. [DOI] [PubMed] [Google Scholar]
  • [56].Lohi J, Wilson CL, Roby JD, Parks WC. Epilysin, a novel human matrix metalloproteinase (MMP-28) expressed in testis and keratinocytes and in response to injury. The Journal of biological chemistry. 2001;276:10134–44. doi: 10.1074/jbc.M001599200. [DOI] [PubMed] [Google Scholar]
  • [57].Werner SR, Mescher AL, Neff AW, King MW, Chaturvedi S, Duffin KL, et al. Neural MMP-28 expression precedes myelination during development and peripheral nerve repair. Developmental dynamics : an official publication of the American Association of Anatomists. 2007;236:2852–64. doi: 10.1002/dvdy.21301. [DOI] [PubMed] [Google Scholar]
  • [58].Chiao YA, Zamilpa R, Lopez EF, Dai Q, Escobar GP, Hakala K, et al. In vivo matrix metalloproteinase-7 substrates identified in the left ventricle post-myocardial infarction using proteomics. Journal of proteome research. 2010;9:2649–57. doi: 10.1021/pr100147r. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].de Castro Bras LE, Cates CA, DeLeon-Pennell KY, Ma Y, Iyer RP, Halade GV, et al. Citrate synthase is a novel in vivo matrix metalloproteinase-9 substrate that regulates mitochondrial function in the postmyocardial infarction left ventricle. Antioxidants & redox signaling. 2014;21:1974–85. doi: 10.1089/ars.2013.5411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].DeCoux A, Lindsey ML, Villarreal F, Garcia RA, Schulz R. Myocardial matrix metalloproteinase-2: inside out and upside down. J Mol Cell Cardiol. 2014;77:64–72. doi: 10.1016/j.yjmcc.2014.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Lindsey ML, Iyer RP, Zamilpa R, Yabluchanskiy A, DeLeon-Pennell KY, Hall ME, et al. A Novel Collagen Matricryptin Reduces Left Ventricular Dilation Post-Myocardial Infarction by Promoting Scar Formation and Angiogenesis. J Am Coll Cardiol. 2015;66:1364–74. doi: 10.1016/j.jacc.2015.07.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Phatharajaree W, Phrommintikul A, Chattipakorn N. Matrix metalloproteinases and myocardial infarction. Can J Cardiol. 2007;23:727–33. doi: 10.1016/s0828-282x(07)70818-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Zamilpa R, Lopez EF, Chiao YA, Dai Q, Escobar GP, Hakala K, et al. Proteomic analysis identifies in vivo candidate matrix metalloproteinase-9 substrates in the left ventricle post-myocardial infarction. Proteomics. 2010;10:2214–23. doi: 10.1002/pmic.200900587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Kirk J. Thrombospondins in the Transition from Myocardial Infarction to Heart Failure. J Mol Cell Cardiol. 2016 doi: 10.1016/j.yjmcc.2015.12.009. in press (JMCC9607) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Chiao YA, Zamilpa R, Lopez EF, Dai Q, Escobar GP, Hakala KW, et al. In vivo Matrix Metalloproteinase-7 Substrates Identified in the Left Ventricle Post-Myocardial Infarction Using Proteomics. Journal of proteome research. 2010;9:2649–57. doi: 10.1021/pr100147r. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Nagase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovascular research. 2006;69:562–73. doi: 10.1016/j.cardiores.2005.12.002. [DOI] [PubMed] [Google Scholar]
  • [67].Dozier S, Escobar GP, Lindsey ML. Matrix metalloproteinase (MMP)-7 activates MMP-8 but not MMP-13. Medicinal chemistry (Shariqah, United Arab Emirates) 2006;2:523–6. doi: 10.2174/157340606778250261. [DOI] [PubMed] [Google Scholar]
  • [68].Van den Steen PE, Proost P, Wuyts A, Van Damme J, Opdenakker G. Neutrophil gelatinase B potentiates interleukin-8 tenfold by aminoterminal processing, whereas it degrades CTAP-III, PF-4, and GRO-alpha and leaves RANTES and MCP-2 intact. Blood. 2000;96:2673–81. [PubMed] [Google Scholar]
  • [69].Teeling JL, De Groot ER, Eerenberg AJ, Bleeker WK, Van Mierlo G, Aarden LA, et al. Human intravenous immunoglobulin (IVIG) preparations degranulate human neutrophils in vitro. Clinical and experimental immunology. 1998;114:264–70. doi: 10.1046/j.1365-2249.1998.00697.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Zhang W, Voice J, Lachmann PJ. A systematic study of neutrophil degranulation and respiratory burst in vitro by defined immune complexes. Clinical and experimental immunology. 1995;101:507–14. doi: 10.1111/j.1365-2249.1995.tb03142.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Banda MJ, Clark EJ, Werb Z. Selective proteolysis of immunoglobulins by mouse macrophage elastase. The Journal of experimental medicine. 1983;157:1184–96. doi: 10.1084/jem.157.4.1184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Heidt T, Courties G, Dutta P, Sager HB, Sebas M, Iwamoto Y, et al. Differential contribution of monocytes to heart macrophages in steady-state and after myocardial infarction. Circ Res. 2014;115:284–95. doi: 10.1161/CIRCRESAHA.115.303567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Tian Y, Koganti T, Yao Z, Cannon P, Shah P, Pietrovito L, et al. Cardiac extracellular proteome profiling and membrane topology analysis using glycoproteomics. Proteomics Clinical applications. 2014;8:595–602. doi: 10.1002/prca.201400009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Tian Y, Zhang H. Glycoproteomics and clinical applications. Proteomics Clinical applications. 2010;4:124–32. doi: 10.1002/prca.200900161. [DOI] [PubMed] [Google Scholar]
  • [75].Lindsey ML, Mayr M, Gomes AV, Delles C, Arrell DK, Murphy AM, et al. Transformative Impact of Proteomics on Cardiovascular Health and Disease: A Scientific Statement From the American Heart Association. Circulation. 2015;132:852–72. doi: 10.1161/CIR.0000000000000226. [DOI] [PubMed] [Google Scholar]
  • [76].Patterson NL, Iyer RP, de Castro Bras LE, Li Y, Andrews TG, Aune GJ, et al. Using proteomics to uncover extracellular matrix interactions during cardiac remodeling. Proteomics Clinical applications. 2013;7:516–27. doi: 10.1002/prca.201200100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].de Castro Bras LE, Deleon KY, Ma Y, Dai Q, Hakala K, Weintraub ST, et al. Plasma fractionation enriches post-myocardial infarction samples prior to proteomics analysis. International journal of proteomics. 2012;2012:397103. doi: 10.1155/2012/397103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Lindsey ML, Weintraub ST, Lange RA. Using extracellular matrix proteomics to understand left ventricular remodeling. Circulation Cardiovascular genetics. 2012;5:o1–7. doi: 10.1161/CIRCGENETICS.110.957803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Holmes J. Modifying the Mechanics of Healing Infarcts: Is Better the Enemy of Good? J Mol Cell Cardiol. 2016 doi: 10.1016/j.yjmcc.2015.11.028. in press (JMCC9569) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Manso AM, Elsherif L, Kang S-M, Ross RS. Integrins, membrane-type matrix metalloproteinases and ADAMs: Potential implications for cardiac remodeling. Cardiovascular research. 2006;69:574–84. doi: 10.1016/j.cardiores.2005.09.004. [DOI] [PubMed] [Google Scholar]
  • [81].Zhang P, Shen M, Fernandez-Patron C, Kassiri Z. ADAMs Family and Relatives in Cardiovascular Physiology and Pathology. J Mol Cell Cardiol. 2015 doi: 10.1016/j.yjmcc.2015.10.031. in press. [DOI] [PubMed] [Google Scholar]
  • [82].Saucerman J. Computational modeling of Cardiac fibroblasts and fibrosis. J Mol Cell Cardiol. 2016 doi: 10.1016/j.yjmcc.2015.11.020. in press (JMCC9598) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Launay G, Salza R, Multedo D, Thierry-Mieg N, Ricard-Blum S. MatrixDB, the extracellular matrix interaction database: updated content, a new navigator and expanded functionalities. Nucleic acids research. 2015;43:D321–7. doi: 10.1093/nar/gku1091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Francis Stuart S, De Jesus N, Lindsey ML, Ripplinger CM. The crossroads of inflammation, fibrosis, and arrhythmia following myocardial infarction. J Mol Cell Cardiol. 2015 doi: 10.1016/j.yjmcc.2015.12.024. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Mann D. Role of inflammatory cells in fibroblast activation. J Mol Cell Cardiol. 2016 doi: 10.1016/j.yjmcc.2015.11.016. in press (JMCC9594) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Turner N. Inflammatory and Fibrotic Responses of Cardiac Fibroblasts to Myocardial Damage Associated Molecular Patterns (DAMPs) J Mol Cell Cardiol. 2016 doi: 10.1016/j.yjmcc.2015.11.002. in press (JMCC9542) [DOI] [PubMed] [Google Scholar]
  • [87].Nahrendorf M. Monocyte and Macrophage Contributions to Cardiac Remodeling. J Mol Cell Cardiol. 2016 doi: 10.1016/j.yjmcc.2015.11.015. in press (JMCC9602) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Johnson JL, Devel L, Czarny B, George SJ, Jackson CL, Rogakos V, et al. A selective matrix metalloproteinase-12 inhibitor retards atherosclerotic plaque development in apolipoprotein E-knockout mice. Arteriosclerosis, thrombosis, and vascular biology. 2011;31:528–35. doi: 10.1161/ATVBAHA.110.219147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Lauer-Fields JL, Whitehead JK, Li S, Hammer RP, Brew K, Fields GB. Selective modulation of matrix metalloproteinase 9 (MMP-9) functions via exosite inhibition. The Journal of biological chemistry. 2008;283:20087–95. doi: 10.1074/jbc.M801438200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Yamamoto D, Takai S. Pharmacological implications of MMP-9 inhibition by ACE inhibitors. Current medicinal chemistry. 2009;16:1349–54. doi: 10.2174/092986709787846514. [DOI] [PubMed] [Google Scholar]
  • [91].Sakata Y, Yamamoto K, Mano T, Nishikawa N, Yoshida J, Hori M, et al. Activation of matrix metalloproteinases precedes left ventricular remodeling in hypertensive heart failure rats: its inhibition as a primary effect of Angiotensin-converting enzyme inhibitor. Circulation. 2004;109:2143–9. doi: 10.1161/01.CIR.0000125741.88712.77. [DOI] [PubMed] [Google Scholar]
  • [92].Lindsey ML, Iyer RP, Zamilpa R, Yabluchanskiy A, DeLeon-Pennell KY, Hall ME, et al. A Novel Collagen Matricryptin Reduces Left Ventricular Dilation Post-Myocardial Infarction by Promoting Scar Formation and Angiogenesis. J Am Coll Cardiol. 2015 doi: 10.1016/j.jacc.2015.07.035. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES