myocardial proteases and matrix remodeling in inflammatory heart disease

11
Review Myocardial proteases and matrix remodeling in inflammatory heart disease Susanne Rutschow, Jun Li, Heinz-Peter Schultheiss, Matthias Pauschinger * Department of Cardiology and Pneumology, Charite, University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany Received 1 June 2005; received in revised form 5 December 2005; accepted 9 December 2005 Time for primary review 22 days Abstract Recently, it has been demonstrated that myocardial inflammation plays a pivotal role in the development and progression of congestive heart failure. The myocardial inflammatory reaction not only affects myocardial hypertrophy and apoptosis, but it has a major influence on the regulation of extracellular matrix turnover. The balance between collagen synthesis and degradation is of crucial relevance in maintaining the structural integrity of the heart. Therefore, the overwhelming inflammatory response, as seen in acute myocarditis or inflammatory cardiomyopathy, could lead to a breakdown of this tightly regulated system. This is an additional key factor in the development and progression of heart failure. This review summarizes the importance of myocardial inflammation in respect to extracellular matrix remodeling and its possible patho- physiological role in the development and progression of left ventricular dysfunction in inflammatory heart disease. D 2006 European Society of Cardiology. Published by Elsevier B.V. Keywords: Myocarditis; DCM; Inflammation; MMP; Matrix 1. Introduction Myocarditis, an inflammatory disorder, which is most commonly caused by viral infection (especially enterovirus and parvovirus), is associated with acute left ventricular dysfunction accompanied by myocardial inflammatory cell infiltration and increased release of proinflammatory cyto- kines [1]. The presence of viral genome in endomyocardial biopsies of patients with dilated cardiomyopathy (DCM) demonstrates an interrelationship between acute myocarditis and DCM [2–4]. A similar frequency of intramyocardial inflammation was found in patients with DCM, which reflects the relevance of chronic inflammation in the development of left ventricular dysfunction [5]. However, not only viral persistence and chronic inflammatory response seem to play an important role in the pathogenesis of inflammatory cardiomyopathy. The latest investigations advert to a momentous role of matrix metalloproteinases in the disruption of the extracellular matrix (ECM) architecture involving a pathological elevated myocardial collagen turnover which leads to the loss of structural integrity of the heart resulting in left ventricular dysfunction [1,6–8]. In the latest investigations these factors have been shown to play a crucial role in the disruption of the ECM architecture. A pathologically elevated myocardial collagen turnover leads to the loss of structural integrity of the heart and ensuing left ventricular dysfunction. Acute left ventricular dysfunction in experimental induced acute myocarditis is associated with induction of proinflammatory cytokines and an imbalance of the matrix metalloproteinases (MMPs) and the tissue inhibitors of MMPs (TIMPs) system [1]. Proinflammatory cytokines like TNF-a and IL-1h contribute not only to depression of LV function and cardiomyocyte loss by apoptosis, they also play a critical role in maintaining the balance in ECM- remodeling [9–11], showing the importance of chronic inflammation in myocardial remodeling process as well as in the development of DCM [12]. They regulate cardiac fibroblast function with the expression of collagen types I and III and fibronectin, and moreover also regulate the 0008-6363/$ - see front matter D 2006 European Society of Cardiology. Published by Elsevier B.V. doi:10.1016/j.cardiores.2005.12.009 * Corresponding author. Tel.: +49 30 8445 2344; fax: +49 30 8445 3565. E-mail address: [email protected] (M. Pauschinger). Cardiovascular Research 69 (2006) 646 – 656 www.elsevier.com/locate/cardiores by guest on June 1, 2013 http://cardiovascres.oxfordjournals.org/ Downloaded from

Upload: fordham

Post on 05-Dec-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

www.elsevier.com/locate/cardiores

Cardiovascular Research

Review

Myocardial proteases and matrix remodeling in inflammatory heart disease

Susanne Rutschow, Jun Li, Heinz-Peter Schultheiss, Matthias Pauschinger *

Department of Cardiology and Pneumology, Charite, University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany

Received 1 June 2005; received in revised form 5 December 2005; accepted 9 December 2005

Time for primary review 22 days

http://cardiovascres.oxfoD

ownloaded from

Abstract

Recently, it has been demonstrated that myocardial inflammation plays a pivotal role in the development and progression of congestive

heart failure. The myocardial inflammatory reaction not only affects myocardial hypertrophy and apoptosis, but it has a major influence on

the regulation of extracellular matrix turnover. The balance between collagen synthesis and degradation is of crucial relevance in maintaining

the structural integrity of the heart. Therefore, the overwhelming inflammatory response, as seen in acute myocarditis or inflammatory

cardiomyopathy, could lead to a breakdown of this tightly regulated system. This is an additional key factor in the development and

progression of heart failure.

This review summarizes the importance of myocardial inflammation in respect to extracellular matrix remodeling and its possible patho-

physiological role in the development and progression of left ventricular dysfunction in inflammatory heart disease.

D 2006 European Society of Cardiology. Published by Elsevier B.V.

rdj

b

ournals.org/

Keywords: Myocarditis; DCM; Inflammation; MMP; Matrix

y guest on June 1, 2013

1. Introduction

Myocarditis, an inflammatory disorder, which is most

commonly caused by viral infection (especially enterovirus

and parvovirus), is associated with acute left ventricular

dysfunction accompanied by myocardial inflammatory cell

infiltration and increased release of proinflammatory cyto-

kines [1]. The presence of viral genome in endomyocardial

biopsies of patients with dilated cardiomyopathy (DCM)

demonstrates an interrelationship between acute myocarditis

and DCM [2–4]. A similar frequency of intramyocardial

inflammation was found in patients with DCM, which

reflects the relevance of chronic inflammation in the

development of left ventricular dysfunction [5]. However,

not only viral persistence and chronic inflammatory

response seem to play an important role in the pathogenesis

of inflammatory cardiomyopathy. The latest investigations

advert to a momentous role of matrix metalloproteinases in

0008-6363/$ - see front matter D 2006 European Society of Cardiology. Publish

doi:10.1016/j.cardiores.2005.12.009

* Corresponding author. Tel.: +49 30 8445 2344; fax: +49 30 8445 3565.

E-mail address: [email protected] (M. Pauschinger).

the disruption of the extracellular matrix (ECM) architecture

involving a pathological elevated myocardial collagen

turnover which leads to the loss of structural integrity of

the heart resulting in left ventricular dysfunction [1,6–8]. In

the latest investigations these factors have been shown to

play a crucial role in the disruption of the ECM architecture.

A pathologically elevated myocardial collagen turnover

leads to the loss of structural integrity of the heart and

ensuing left ventricular dysfunction.

Acute left ventricular dysfunction in experimental

induced acute myocarditis is associated with induction of

proinflammatory cytokines and an imbalance of the matrix

metalloproteinases (MMPs) and the tissue inhibitors of

MMPs (TIMPs) system [1]. Proinflammatory cytokines like

TNF-a and IL-1h contribute not only to depression of LV

function and cardiomyocyte loss by apoptosis, they also

play a critical role in maintaining the balance in ECM-

remodeling [9–11], showing the importance of chronic

inflammation in myocardial remodeling process as well as

in the development of DCM [12]. They regulate cardiac

fibroblast function with the expression of collagen types I

and III and fibronectin, and moreover also regulate the

69 (2006) 646 – 656

ed by Elsevier B.V.

S. Rutschow et al. / Cardiovascular Research 69 (2006) 646–656 647

matrix degradation system by influencing the expression of

matrix metalloproteinases (MMPs), the tissue inhibitors of

MMPs (TIMPs) and their activators like urokinase type

plasminogen activator and tissue type plasminogen activator

(uPA, tPA) [12–15]. This seems to indicate that the

maintenance of the physiological myocardial matrix turn-

over involves a highly regulated interaction between cardiac

and noncardiac cells, in response to the release of

inflammatory mediators and components of the matrix

degradation system.

Fig. 1. (a) Significant correlation between myocardial mRNA abundance of

IL-1h and left ventricular endsystolic pressure (LVESP) in a murine model

of Coxsackievirus B3 induced acute myocarditis. R2>0.5 were considered

as significant; p <0.001. (b) Significant correlation between myocardial

mRNA abundance of TNF-a and left ventricular endsystolic pressure

(LVESP) in a murine model of Coxsackievirus B3 induced acute

myocarditis. R2>0.5 were considered as significant; p <0.001.

by guest on June 1, 2013http://cardiovascres.oxfordjournals.org/

Dow

nloaded from

2. Heart failure and cytokines

Chronic heart failure is a disease with multiple causes of

which an inflammatory reaction is the most important. Over

the past years inflammatory cytokines and neurohormones

have been shown to contribute to the development and

progression of left ventricular dysfunction. Congestive heart

failure is now commonly seen as a state of chronic

inflammation. Proinflammatory cytokines like TNF-a and

IL-6 are elevated in different entities of heart failure like

acute myocardial infarction, stable angina pectoris or acute

myocarditis with perpetuation of congestive heart failure

and relevance for prognosis of the disease [16–19]. For

instance, TNF-a has been shown to induce myocardial

hypertrophy and fibrosis [20], increase cardiac myocyte

apoptosis and induce activation of the inducible form of

nitric oxide synthase (iNOS) [21]. IL-1 has been demon-

strated to depress myocardial contractility in a dose-

dependent manner and to contribute to myocardial apoptosis

and hypertrophy. In collaboration with IL-1h, TNF-a also

promotes Coxsackievirus B3-myocarditis in resistant Balb/

c-mice [22]. In our previous investigations we could show a

close correlation between the development of left ventric-

ular dysfunction and the increase of myocardial mRNA

abundance of IL-1h and TNF-a in a murine model of CVB-

3 induced acute myocarditis (Fig. 1). Furthermore, Liu and

Zhao have demonstrated that heart function is improved by

declining circulating TNF-a concentration [23]. The neuro-

humoral activation and the elevated oxidative stress during

the development of left ventricular dysfunction in conges-

tive heart failure can be triggered by proinflammatory

cytokines. Increased free oxidative radicals are able to lead

to an activation of p38-MAP-kinase and nuclear factor

kappa B (NFnB). These directly affect left ventricular

dysfunction by inducing cardiomycytolysis and by the

negative inotropic effects of reduced calcium uptake by

the sarcoplasmatic reticulum [24,25]. Another important

pathophysiologic process in the development of congestive

heart failure concerns the sympathic nervous system with

increased levels of catecholamines, which may result in an

overwhelming inflammatory reaction. Murray et al. have

demonstrated that chronic h-adrenergic stimulation leads to

an increase of myocardial gene expression and protein

production of TNF-a, IL-1h and IL-6 [26]. This result

confirms previous studies showing that isoproterenol

advances myocardial hypertrophy [27], myocyte degenera-

tion and inflammatory cell infiltration [28]. This is also in

line with our investigation, as described below, which shows

that the treatment of acute myocarditis with a h-adrenergicreceptor blocker leads to a reduced inflammatory response,

modulated immune reaction and decreased ECM remodel-

ing as well as an improved left ventricular function [29].

S. Rutschow et al. / Cardiovascular Research 69 (2006) 646–656648

by guest on June 1, 2013http://cardiovascres.oxfordjournals.org/

Dow

nloaded from

3. Myocarditis and dilated cardiomyopathy

Myocarditis, an inflammatory disease of the myocardi-

um, diagnosed by standardized histological and immuno-

histological methods [30], is caused by both infectious and

noninfectious agents [31]. However, the main cause is the

viral infection of the myocardium with cardiotropic viruses,

like enterovirus or parvovirus [32]. Previous investigations

on endomyocardial biopsies showed that several other viral

agents like adenovirus or parvovirus B19 are also involved

in the etiology of myocarditis [3,4]. For the pathogenesis of

myocarditis, Kawai described a triphasic model with an

acute, subacute and chronic phase [33]. During the acute

phase the myocardium mainly expresses proinflammatory

cytokines, such as IL-1h, TNF-a and Interferon-g [34,35].

These elevated proinflammatory cytokine levels are linked

to histological changes such as myocyte necrosis and

apoptosis [36,37]. In this early stage of disease there is

very little infiltration of natural killer cells and macrophages

in the myocardium [38,39]. This phase is characterized by a

dominance of direct pathogenicity of viral action and

myocyte apoptosis with direct alteration of myocardial

architecture by destroying dystrophin [40–43]. In the

subacute phase an increase in myocardial cell infiltration,

especially T-lymphocytes, natural killer cells and fibroblasts

follows [33]. The protective effect of natural killer cells is

based on their suppression of viral replication by the

perforin-induced disturbance of infected cardiomyocytes

[44,45]. The importance of early suppression of viral

replication is shown by the more severe myocarditis seen

in a murine myocarditis model with a defect of NK-cells

[46]. Seko et al. could detect a time-dependent release of

cytokines, with an initial increase of proinflammatory

cytokines (IL-1h, IL-6, TNF-a, Interferon-g) to the 7th

day after infection, followed by regulatory cytokines like

IL-2, IL-4 and IL-10 [47]. The outcome of the virus-

induced immune response is not straightforward and may

vary greatly. An efficient viral elimination through a radical

immune response is required for a better outcome of the

disease. For example, patients with myocarditis who have

higher cardiac specific IgG have better survival prospects

[48] and furthermore, acute fulminant myocarditis with an

initial massive inflammation and more pronounced cellular

injury has a better long-term outcome as compared with

acute nonfulminant myocarditis [49]. On the other hand an

uncontrolled immune response is associated with an

inefficient viral elimination, pathological myocardial in-

flammation, and initiating of cross-reacting antibodies,

which result in progressive cellular injury and matrix

remodeling. The increase of cardiac cell-infiltration and

release of inflammatory cytokines directly correlate to the

development of left ventricular dysfunction [50]. Viral

persistence and chronic inflammation may finally lead to

the third and chronic phase of myocarditis with the

development of dilated cardiomyopathy. Enteroviral ge-

nome could be detected in the myocardium up to 90 days

after viral infection in a murine model of myocarditis,

which indicates one causal step of chronic myocardial cell

infiltration [51,52]. The importance of latent or persistent

virus with induced chronic myocardial inflammation has

been demonstrated in persistent Cytomegalovirus-myocar-

ditis [53] and in the development of left ventricular

dilatation in mice with persistence of Coxsackievirus B3

[54]. The presence of viral genome in endomyocardial

biopsies of patients with dilated cardiomyopathy underlines

the relationship between myocarditis and the development

of left ventricular dysfunction [2]. Another important step in

the development and progression of acute left ventricular

dysfunction and dilatation is the initiation of an adverse

remodeling of the ECM by immune mediators. Chronic

myocardial inflammation induces the pathological collagen

turnover resulting in loss of structural integrity of the

myocardium and in the development of left ventricular

dysfunction.

4. The extracellular matrix—collagen and cytokines

Myocardial ECM (mainly collagen type I and III and

fibronectin, elastin, etc.) forms a three dimensional cross-

linked collagen-network which supports the cellular and

structural integrity of the heart. After secretion into the

ECM, the fibril-forming collagens aggregate spontaneously

after following the processing of procollagens into ordered

fibrillar network, stabilized by covalent cross-links. The

structural elements are dynamic and there is a complex

network of receptors and enzymes within the ECM which

controls the turnover of this tightly regulated system.

Collagen production and degradation is an important

process in the development of left ventricular dysfunction

in acute and chronic myocarditis and determines the

maintenance of cardiac architecture.

Collagen synthesis by cardiac fibroblasts is regulated by

multiple factors, including inflammatory cytokines like

TNF-a, IL-1h or TGF-h, TNF-a and IL-1h have been

shown to decrease the expression of procollagen type I and

III and increase the expression of fibronectin and non-

fibrillar procollagen type IV. Furthermore, several other

factors like aldosterone, TGF-h or mechanical stretch have

been shown to induce the mRNA synthesis of collagen [12].

The renin-angiotensine-system also partakes in the regula-

tion of collagen synthesis; angiotensin II induces ECM

protein synthesis and accumulation via AT1-receptor stim-

ulation, effects that are mediated by TGF-h1 and endothelin-

1 [55]. A rat model of myocardial infarction showed the

importance of this myocardial remodeling: ACE inhibition

prevented collagen accumulation and DNA synthesis and

furthermore completely inhibited collagen deposition with

AT1-receptor antagonism [56].

Fibrillar collagen turnover results from the equilibrium

between the synthesis and the degradation of collagen,

mainly types I and III. In the healthy left ventricle ECM

S. Rutschow et al. / Cardiovascular Research 69 (2006) 646–656 649

replacement is about 5% to 9%, in pathological conditions,

e.g. after myocardial infarction, this figure can rise to 50%

[57]. Furthermore the rate of collagen synthesis per day is

much slower than of noncollagen proteins with a longer

half-life period of collagen, implying a fairly slow ECM

replacement after degradation and potential vulnerability for

adverse remodeling [58]. This additionally reduces the

ability to form the three dimensional network with collagen

cross-links, which leads to alteration in the composition and

structure of myocardial collagen.

by guehttp://cardiovascres.oxfordjournals.org/

Dow

nloaded from

5. The MMP system

Collagen degradation is an important step in ECM

remodeling, mainly due to matrix metalloproteinases

(MMPs) and serine-proteinases. The matrix metalloprotei-

nases are a family of 20 different species of zinc-dependent

enzymes [59], which could be expressed under basal

conditions by fibroblasts and myocytes and also in response

to inflammatory reactions by infiltrating macrophages and

lymphocytes. One group of MMPs is secreted into the

extracellular space in a latent or proenzyme form and the

other group is membrane-bound. Regarding the catalytic

domain of MMPs, a large extracellular binding domain at the

C-terminus is responsible for the substrate specificity and the

specific binding to ECM proteins [59,60]. In addition, these

substrate specificities and functions classify the secreted

MMPs in three different groups. First the collagenases

(MMP-1, MMP-8, MMP-13) decompose the insoluble

collagen fibrils into soluble fragments [61]. The cleaving

of these soluble fragments is continued by gelatinases

(MMP-2, MMP-9), which are highly expressed in the left

Fig. 2. Simplified mechanism of regulation in the Matrix-Degr

ventricular myocardium [7,62]. Coker et al. showed an

isolated expression of these gelatinases in LV-myocytes,

which supports the possibility of a direct processing of

matrix remodeling by the synthesis and release of MMPs

[63]. MMP-2 is thereby constitutively expressed in the

myocardium, whereas MMP-9 is an inducible form of the

MMPs and becomes more relevant under inflammation with

inducible expression in neutrophils and macrophages

[64,65]. The third group, the stromelysine-like MMP-3,

degrades not only a wide range of components of the ECM, it

can also initiate the activation of the inactive pro-MMPs.

The group of membrane-bound MMPs (Membrane

Type-MMP, MT-MMP) plays also a critical role in matrix

remodeling, which on the one hand cleaves intact fibrillar

collagen and basement membrane components and on the

other hand directly activates different pro-MMPs [66].

However MMPs do not only play a role in the degradation

of the ECM-components, the final fragmented matrix

peptides also have biological activities and have been

shown to stimulate new collagen synthesis by cardiac

fibroblasts [67].

Beside the ECM components, the target substrates of the

MMPs also include nonmatrix proteins such as cytokines,

receptors and adhesions molecules [68–70]. This explains

additional functions of MMPs as for example regulating

growth, cell pathways, and angiogenesis [71]. The capabil-

ity of activated MMPs to degrade the complete ECM is

conditioned by a tightly controlled system. First, the control

at the transcriptional level by several cytokines or growth

factors; second the control of pro-MMP activation by serine

proteinases (Plasmin-system), MT-MMPs or stromelysines;

and last the inhibition of activated MMPs by the tissue

inhibitors of MMPs (TIMP1-4) (Fig. 2).

adation-System. Importance of inflammatory mediators.

st on June 1, 2013

S. Rutschow et al. / Cardiovascular Research 69 (2006) 646–656650

by guest on June 1, 2013http://cardiovascres.oxfordjournals.org/

Dow

nloaded from

5.1. Synthesis of MMPs

The synthesis of MMPs at the transcriptional level is

influenced by multiple cytokines, neurohormones and

growth factors [72]. Different signaling pathways have

been shown to be involved in regulation of the expression of

MMPs [73–80].

It could be demonstrated that proinflammatory cytokines

like IL-1h, TNF-a or IL-6 induce MMP-synthesis [12,13].

Furthermore, in mice with over expressing TNF-a, a

progressive development of left ventricular dilatation is

associated with increased MMP-activity [20]. In collabora-

tion with TNF-a, interferon-g enhances the MMP-1

synthesis in human monocytes and neutralizing antibodies

against TNF-a block the induction of MMP-1 by interferon-

g [81]. By contrast, TGF-h1 on the one hand decreases the

proteolytic activity of MMP-1 and MMP-3 by suppressing

MMP gene expression through the TGF-h inhibitory

element (TIE) and on the other hand increases the

expression of MMP-2 and MMP-9 [14,82]. Beside inflam-

matory cytokines, several other bioactive molecules also

influence the synthesis of MMPs. Angiotensin II plays a

pivotal role here: It has been demonstrated that angiotensin

II induces the expression of MMP-2, -9 and -14 in neonatal

rat fibroblast [83]. Its importance in this regulating process

is further shown by an improvement in left ventricular

function and myocardial geometry through reduced MMP-

synthesis after ACE-inhibition or AT-1 blockade [84,85].

Endothelin-1 enhances the production of MMP-2 and

MMP-9 and endothelin receptor blockade results in reduced

levels of MMP-2 and -9 [86]. Furthermore, natriuretic

peptides like BNP (brain natriuretic peptide) are able to

induce the MMP-synthesis in chronic heart failure and

myocardial remodeling [87].

5.2. Activation of MMPs

MT-MMPs and MMP-3 activate different members of

the MMP family by proteolytic cleavage of the MMP-

propeptide [15,66,88]. Even more important is the plasmin-

ogen system (plasminogen activators (PA), tissue-type PA

(t-PA), urokinase-type PA (uPA) and the PA-inhibitor

(PAI)), which activates the inactive proMMPs as described

above [88]. The importance of this system in disrupting the

cardiac collagen network has been previously demonstrated

in mice lacking uPA or mice with acute pressure overload

with PAI gene transfer showing improved left ventricular

function with reduced myocardial fibrosis and preserved

interstitial matrix [89]. Beside MMP-activation the PA are

also able to cleave matrix proteins like fibrin and

fibronectin. The expression of uPA [90], which has the

same transcription factor binding elements (AP-1, PEA3) in

the promotor region like MMP-1 and MMP-3 is induced by

IL-1h and TNF-a [91]. In fibroblasts, IL-1h increases not

only the protein and mRNA expression of uPA but also its

receptor uPAR [92]. Furthermore, there is a feedback

regulation between the MMPs and the plasmin-system;

MMP-3 has been demonstrated to cleave their physiological

inhibitor (PAI, a2-antiplasmin) resulting in neutralization

[93,94].

5.3. Inhibition of MMPs

Active MMPs are specifically inhibited by their endog-

enous inhibitors, the TIMPs that are composed of four

subtypes (TIMP-1-4) [81,95–97]. TIMPs bind to the active

site of MMP in a 1:1 stoichiometrie, blocking their access to

collagen substrate [15,98]. Furthermore they can bind latent

MMPs at the aminoterminus thereby preventing their

activation [99]. Some differences in the inhibitory properties

of the different TIMPs have been reported. TIMP-2 and

TIMP-3 have been proven to be effective inhibitors of

membrane-bound-MMPs, and TIMP-3 is the only TIMP

which can inhibit TNF-a converting enzyme [100,101].

Mice deficient of TIMP-1 gene develop increased left

ventricular mass and increased end diastolic volume

suggesting its important role in the prevented activation of

the protease cascade and in the maintenance of a dynamic

matrix balance. Inflammatory mediators also influence this

third step in the control of MMP-activity; IL-1 and TNF-a

can down regulate the expression of TIMP-1 [15]. Beside

this specific inhibition a2 macroglubulin and heparin

nonspecifically inhibit MMP-activity [102,103].

This widespread influence of inflammatory reaction on

the regulation of the matrix degradation system reflects a

deregulation in disease with pathologically elevated inflam-

matory mediators.

6. Extracellular matrix remodeling

6.1. ECM in acute myocarditis

Our previous investigations focused on the inflammatory

reaction in virus-induced acute myocarditis caused by

disruption of the collagen turnover and the resulting

changes in the ECM and its influence on left ventricular

dysfunction [1]. We could show that in the early phase of

myocarditis the main disruption of ECM is caused by

qualitative changes in the collagen network. Expression of

myocardial mRNA and protein abundance of collagen type

I was unchanged as was total collagen content measured by

picrosirius red staining. However Western blot analysis

demonstrated an increased fraction of soluble collagen,

suggesting an initial dominance of posttranslational colla-

gen variation contingent on an imbalance in the matrix

degradation system. Corroborating these findings, Wood-

iwiss et al. have demonstrated that a reduction in collagen

cross-links is responsible for the decreased native insoluble

or increased soluble collagen and is associated with left

ventricular dysfunction, remodeling and chamber dilatation

[104]. MMP-9, which depolimerizes the cross-linked

S. Rutschow et al. / Cardiovascular Research 69 (2006) 646–656 651

http://cardiovascres.oxfD

ownloaded from

polymers of collagen type I [105], seems to play an

important role. Furthermore, LV enlargement after myocar-

dial infarction could be prevented by deletion of the MMP-

9 gene [106]. CVB-3 infection of the myocardium caused a

significant release of proinflammatory cytokines such as IL-

1h, TNF-a and TGF-h on the 10th day post infection. This

was accompanied by an up regulated expression of MMP-3

and MMP-9 and reduced levels of their endogenous specific

inhibitors TIMP-1 and TIMP-4. Furthermore, our unpub-

lished data show evidence of an imbalance in the plasmin

regulation system in this acute early phase of myocarditis

by significant induced mRNA abundance of uPA with

simultaneously reduced levels of PAI mRNA abundance

(Fig. 3). As described above, a restored interstitial matrix

with an undilated chamber could be demonstrated in a uPA

knock-out model of chronic volume overload [89], suggest-

ing that the above demonstrated imbalance in the matrix

degradation system was mainly responsible for the devel-

opment of left ventricular dysfunction. Increased levels of

MMPs, decreased levels of TIMPs and the activated

plasmin system in the acute phase of myocarditis may lead

to an imbalance in the matrix degradation system in favour

of matrix protein degradation. This ultimately reduces the

matrix integrity and disrupts the three dimensional collagen

network by cleaving the cross-links between the collagen

molecules, which then leads to left ventricular dysfunction

and dilatation.

Fig. 3. Differential mRNA abundances of myocardial MMP-3, TIMP-1, PAI and

Dominance of matrix degrading components with loss of their controlling agents

6.2. ECM in dilated cardiomyopathy

As described above, previous investigations suggested an

association between myocarditis and inflammatory cardio-

myopathy caused by viral persistence. Signs are dystrophin

degradation, myocardial inflammation with cytotoxic T-

lymphocyte-mediated myocytolysis, B-lymphocyte-mediat-

ed generation of autoantibodies and induction of the MMP-

system with pathologic collagen degradation and left

ventricular dilatation. The results of our recent analysis of

endomyocardial biopsies emphasize the importance of

persistent inflammation in dilated cardiomyopathy regarding

ECM alterations and the development of left ventricular

dysfunction. The myocardial mRNA abundance of MMP-3

and TIMP-1 was analyzed in endomyocardial biopsies of

patients with dilated cardiomyopathy with or without

inflammation and in biopsies of patients with normal left

ventricular function without histological signs of inflamma-

tion. We could demonstrate a significantly induced expres-

sion of myocardial MMP-3 in inflammatory cardiomyopathy

accompanied by a reduced expression of TIMP-4 [107] in

dilated cardiomyopathy without inflammation. This is in line

with the results of Schwartzkopff et al. who have shown

elevated serum markers of collagen degradation accompa-

nied by increased levels of MMP-1 in patients with DCM

[108]. Furthermore there was a negative correlation between

the MMP/TIMP ratio and the degree of LV-dilatation,

uPA and gelatinase activity in acute myocarditis on 10th day postinfection.

. Tp <0.05 were considered as significant.

by guest on June 1, 2013ordjournals.org/

S. Rutschow et al. / Cardiovascular Research 69 (2006) 646–656652

by guest on June 1, 2013http://cardiovascres.oxfordjournals.org/

Dow

nloaded from

underlining the importance of collagen degradation in the

development of left ventricular dysfunction. Moreover, we

could detect a co-localization of CD-3 and MMP-3,

measured by immune histochemistry, which further empha-

sizes the importance of inflammation in this pathogenesis.

The increased production of collagen in the later stage of

myocarditis seems to be a response to collagen degradation

caused by increased MMP-activity. Since matrikines en-

hance the synthesis of procollagen it might be possible that

after the initial importance of degradation, fibrosis takes

over in the later phase of myocarditis [58]. This was also

verified by Pauschinger et al. in patients with DCM, who

had an increase in the myocardial collagen content

measured by picrosirius red staining [8]. McCormick et al.

speculated that there was a defect in collagen cross-linking

pathways, leading to an increase in collagen concentration

and collagenase activity with ventricular dilatation since in

patients with idiopathic dilated cardiomyopathy, collagen

deposition was doubled but hydroxyproline concentration

was reduced [109,110]. Furthermore a prospective study on

serum markers of collagen metabolism in idiopathic DCM

showed that patients with increased serum markers had an

increased risk for transplantation, impaired left ventricular

function, or advanced clinical stage of heart failure, and

death suggesting that increased collagen synthesis and

degradation act as a factor in risk stratification [111].

During the fibrotic process, the collagen accumulation

was also accompanied by a change in the ratio of Collagen I

and III, suggesting increased myocardial stiffness with

impaired systolic and diastolic function [8].

Kuhl et al. have demonstrated an improved left

ventricular function by eliminating viral persistence (Ade-

novirus and Enterovirus) through therapy with Interferon-h[112]. In this study the viral elimination led to significantly

reduced inflammation, which furthermore reflects the

pivotal role of viral persistence and chronic induced

inflammation. Kuhl et al. also suggest that active viral

replication interferes with inflammatory cytokine release as

well as with matrix integrity. This may be partially

reversible after viral elimination and could explain im-

proved cardiac function after viral elimination. Further

analysis will show whether the possibly reduced patholog-

ical alterations of the MMP/TIMP system in response to

myocardial collagen deposition play a role in the develop-

ment of improved left ventricular dysfunction after viral

elimination through h-interferon.

6.3. ECM-remodeling in acute myocarditis with

b-adrenergic-blockage

In order to explore the influence of induced sympathic

activity and oxidative stress on cardiac inflammatory

reaction we investigated the effect of a h-adrenergic-blockage on a possible improvement of myocardial injury

and imbalance in the MMP/TIMP-system [29]. The effects

of carvedilol and metoprolol on myocardial inflammation,

ECM remodeling and left ventricular function were

examined in hearts of Coxsackievirus B3 infected Balb/c

mice on the 10th day after infection. Carvedilol could

improve left ventricular dysfunction with a significant

enhancement of LVESP, dP / dtmax and dP / dtmin [113].

This improvement was accompanied by a reduced inflam-

matory reaction as shown by the reduced release of

proinflammatory cytokines such as IL-1h, TNF-a and

TGF-h. The importance of proinflammatory cytokines in

the development of left ventricular function was demon-

strated by a significant correlation between the myocardial

enlargement caused by proinflammatory cytokines and

cardiac hemodynamic parameters. We could furthermore

show an improved balance of the MMP/TIMP-system with

reduced levels of myocardial mRNA abundance of MMPs,

especially MMP-8, reflecting a reduced myocardial cell

infiltration by the cell specific expression of neutrophils

underlined by its significant correlation to the regulation of

IL-1h, TNF-a. This emphasizes the complex interaction of

ECM remodeling, neurohumoral activation, reactive oxygen

species, myocardial cell infiltration and release of proin-

flammatory cytokines. A comparison between carvedilol

and metoprolol showed that carvedilol exerts a better effect

in the improvement of left ventricular function. This might

be due to the absent effect of the selective h-adrenergicblocker metoprolol on the release of myocardial proin-

flammatory cytokines and MMPs. Nishio et al. demonstrat-

ed that the actions of epinephrine could be blocked by

carvedilol and propanolol, but not by metoprolol, suggest-

ing maintenance of epinephrine effects over h2-adrenergic

receptor stimulation [114]. Besides antioxidative properties

[115], carvedilol has also been proven to inhibit the

generation of free oxygen radicals by neutrophils [116]

and to reduce the production of oxidized low density

lipoproteins [117]. This might partially explain its expanded

effects on the MMP/TIMP-system which could explain the

improved cardiac function.

7. Conclusion

The myocardial ECM is a complex network which

balance determines the structural integrity of the heart.

Alteration in the matrix degradation system caused by

inflammatory mediators, oxygen species and neurohumoral

reaction leads to an impairment of left ventricular function

as seen in myocarditis and inflammatory cardiomyopathy.

The imbalance of the matrix degrading system with induced

expression of MMPs and plasminogen activators as well as

the reduced expression of TIMPs, leads to a pathologic

collagen turnover, with the loss of structural integrity of the

heart and an impairment of LV function. Therefore, the

regulation of the MMP/TIMP system is an important

therapeutic target in the prevention of the progression of

inflammatory heart failure. Further investigations are

needed to determine the best target for intervention in order

S. Rutschow et al. / Cardiovascular Research 69 (2006) 646–656 653

to influence this complex system leading to a dynamic

balance between collagen accumulation and degradation.

Acknowledgement

This research was supported by a grant of the Deutsche

Forschungsgemeinschaft (DFG Transregio 19).

by guest on June 1, 2013http://cardiovascres.oxfordjournals.org/

Dow

nloaded from

References

[1] Li J, Schwimmbeck PL, Tschope C, Leschka S, Husmann L,

Rutschow S, et al. Collagen degradation in a murine myocarditis

model: relevance of matrix metalloproteinase in association with

inflammatory induction. Cardiovasc Res 2002;56:235–47.

[2] Kuhl U, Pauschinger M, Noutsias M, Bock T, Lassner D, Poller W, et

al. High prevalence of viral genomes and multiple viral infections in

the myocardium of adults with ‘‘idiopathic’’ left ventricular dysfunc-

tion. Circulation 2005;111:887–93.

[3] Pauschinger M, Bowles NE, Fuentes-Garcia FJ, Pham V, Kuhl U,

Schwimmbeck PL, et al. Detection of adenoviral genome in the

myocardium of adult patients with idiopathic left ventricular

dysfunction. Circulation 1999:1348–54.

[4] Kuhl U, Pauschinger M, Bock T, Klingel K, Schwimmbeck CP,

Seeberg B, et al. Parvovirus B19 infection mimicking acute

myocardial infarction. Circulation 2003;108:945–50.

[5] Noutsias M, Seeberg B, Schultheiss HP, Kuhl U. Expression of cell

adhesion molecules in dilated cardiomyopathy: evidence for endo-

thelial activation in inflammatory cardiomyopathy. Circulation 1999;

99:2124–31.

[6] D’Armiento J. Matrix metalloproteinase disruption of the extracellu-

lar matrix and cardiac dysfunction. Trends Cardiovasc Med 2002;

12:97–101.

[7] Thomas CV, Coker ML, Zellner JL, Handy JR, Crumbley AJ 3rd,

Spinale FG. Increased matrix metalloproteinase activity and selective

upregulation in LV myocardium from patients with endstage dilated

cardiomyopathy. Circulation 1998;97:1708–15.

[8] Pauschinger M, Knopf D, Petschauer S, Doerner A, Poller W,

Schwimmbeck PL, et al. Dilated cardiomyopathy is associated with

significant changes in collagen type I / III ratio. Circulation 1999;99:

2750–6.

[9] Torre-Amione G, Kapadia S, Lee J, Durand JB, Bies RD, Young JB,

et al. Tumor necrosis factor-a and tumor necrosis factor receptors in

the failing human heart. Circulation 1996;93:704.

[10] Torre-Amione G, Kapadia S, Lee J, et al. Expression and functional

significance of tumor necrosis factor receptors in human myocardi-

um. Circulation 1995;92:1487–93.

[11] Muegge K, Williams TM, Kant J, Karin M, Chiu R, Schmidt A, et al.

Interleukin-1 costimulatory activity on the interleukin-2 promoter via

AP-1. Science 1989;246:249–51.

[12] Siwik DA, Chang DL, Colucci WS. Interleukin-1 h and tumor

necrosis factor-a decrease collagen synthesis and increase matrix

metalloproteinase activity in cardiac fibroblasts in vitro. Circ Res

2000;86:1259–65.

[13] Deswal A, Petersen NJ, Feldman AM, White BG, Mann DL.

Cytokines and cytokine receptors in advanced heart failure: an

analysis of the cytokine database from the Vesnarinone trial (VEST).

Circulation 2001;103:2055–9.

[14] Mauviel A. Cytokine regulation of metalloproteinase gene expres-

sion. J Cell Biochem 1993;53:288–95 [Review].

[15] Nagase H. Activation mechanisms of matrix metalloproteinases. Biol

Chem 1997;378:151–60.

[16] Torre-AmioneG,Kapadia S, Benedict C, OralH,Young JB,MannDL.

Proinflammatory cytokine levels in patients with depressed left

ventricular ejection fraction: a report from the Studies of Left

Ventricular Dysfunctions (SOLVD). J Am Coll Cardiol 1996;

27/5:1201–6.

[17] Matsumori A. Cytokines in myocarditis and cardiomyopathies. Curr

Opin Cardiol 1996;11:302–9.

[18] Ikonomidis I, Andreotti F, Economou E, Stefanadis C, Toutouzas P,

Nihoyannopoulos P. Increased proinflammatory cytokines in patients

with chronic stable angina and their reduction by aspirin. Circulation

1999;100:793–8.

[19] Ridker PM, Rifai N, Pfeffer M, Sacks F, Lepage S, Braunwald E.

Elevation of tumor necrosis factor-a and increased risk of recurrent

coronary events after myocardial infarction. Circulation 2000;101:

2149–53.

[20] Sivasubramanian N, Coker ML, Kurrelmeyer KM, MacLellan WR,

DeMayo FJ, Spinale FG, et al. Left ventricular remodeling in

transgenic mice with cardiac restricted overexpression of tumor

necrosis factor. Circulation 2001;104:826–31.

[21] von Haehling S, Jankowska EA, Anker SD. Tumour necrosis factor-

a and the failing heart: pathophysiology and therapeutic implica-

tions. Basic Res Cardiol 2004;99:18–28.

[22] Lane JR, Neumann DA, Lafond-Walker A, Herskowitz A, Rose NR.

Role of IL-1 and tumor necrosis factor in coxsackie virus-induced

autoimmune myocarditis. J Immunol 1993;151:1682–90.

[23] Liu L, Zhao SP. The changes of circulating tumor necrosis factor

levels in patients with congestive heart failure influenced by therapy.

Int J Cardiol 1999;69:77–82.

[24] Liao F, Andalibi A, Qiao JH, Allayee H, Fogelman AM, Lusis AJ.

Genetic evidence for a common pathway mediating oxidative stress,

inflammatory gene induction, and aortic fatty streak formation in

mice. J Clin Invest 1994;94:877–84.

[25] Barnes PJ, Karin M. Nuclear factornB: a pivotal transcription factor

in chronic inflammatory diseases. N Engl J Med 1997;336:1066–71.

[26] Murray DR, Prabhu SD, Chandrasekar B. Chronic h-adrenergicstimulation induces myocardial proinflammatory cytokine expres-

sion. Circulation 2000;101:2338–41.

[27] Boluyt MO, Long X, Eschenhagen T, Mende U, Schmitz W, Crow

MT, et al. Isoproterenol infusion induces alterations in expression of

hypertrophy-associated genes in rat heart. Am J Physiol 1995;269:

H638–47.

[28] Chappel CI, Rona G, Balazs T, Gaudry R. Severe myocardial

necrosis produced by isoproterenol in the rat. Arch Int Pharmacodyn

Ther 1959;122:123–8.

[29] Pauschinger M, Rutschow S, Chandrasekharan K, Westermann D,

Weitz A, Peter Schwimmbeck L, et al. Carvedilol improves left

ventricular function in murine Coxsackievirus-induced acute myo-

carditis association with reduced myocardial interleukin-1h and

MMP-8 expression and a modulated immune response. Eur J Heart

Fail 2005;7:444–52.

[30] Richardson P, McKenna W, Bristow M. Report of the 1995 World

Health Organisation/International Society and Federation of Cardi-

ology task force on the definition and classification of cardiomyop-

athies. Circulation 1996;93:341–2.

[31] O’Connell JB. Diagnosis and medical treatment of inflammatory

cardiomyopathy. In: Topol EJ, editor. Textbook of Cardiovascular

Medicine. Philadelphia’ Lippincott-Raven; 1998. p. 2309–36.

[32] Pauschinger M, Doerner A, Kuehl U, Schwimmbeck PL, Poller W,

Kandolf R, et al. Enteroviral RNA replication in the myocardium of

patients with left ventricular dysfunction and clinically suspected

myocarditis. Circulation 1999:889–95.

[33] Kawai C. From myocarditis to cardiomyopathy: mechanisms of

inflammation and cell death. Learning from the past to the future.

Circulation 1999;96:3549–54.

[34] Shioi T, Matsumori A, Sasayama S. Persistent expression of cytokine

in the chronic stage of viral myocarditis in mice. Circulation

1996;94:2930–7.

[35] Matsumori A. Molecular and immune mechanisms in the pathogen-

esis of cardiomyopathy: role of viruses, cytokines and nitric oxide.

Jpn Circ J 1997;61:275–91.

S. Rutschow et al. / Cardiovascular Research 69 (2006) 646–656654

by guest on June 1, 2013http://cardiovascres.oxfordjournals.org/

Dow

nloaded from

[36] Rose NR, Hill SL. The pathogenesis of postinfectious myocarditis.

Clin Immunol Immunopathol 1996;80:92–9.

[37] Wilson FM, Miranda QR, Chason JL, Lerner M. Residual pathologic

changes following murine coxsackie A and B myocarditis. Am J

Pathol 1969;55:253–65.

[38] Seko Y, Shinkai Y, Kawasaki A, Yagita H, Okumura K, Takaku F, et

al. Expression of perforin in infiltrating cells in murine hearts with

acute myocarditis caused by coxsackievirus B3. Circulation 1991;84:

788–95.

[39] Feldman AM, McNamara D. Myocarditis. N Engl J Med 2000(Nov

9);343:1388–98.

[40] Chow LH, Beisel KW, McManus BM. Enteroviral infection of mice

with severe combined immunodeficiency. evidence for direct viral

pathogenesis of myocardial injury. Lab Invest 1992;66:24–31.

[41] Herzum M, Ruppert V, Kuytz B, Jomaa H, Nakamura I, Maisch B.

Coxsackievirus B3 infection leads to cell death of cardiac myocytes.

J Mol Cell Cardiol 1994;26:907–13.

[42] McManus BM, Chow LH, Wilson JE, Anderson DR, Gulizia JM,

Gauntt CJ, et al. Direct myocardial injury by enterovirus: a central

role in the evolution of murine myocarditis. Clin Immunol

Immunopathol 1993;68:159–69.

[43] Badorff C, Lee GH, Lamphear BJ, Martone ME, Campbell KP,

Rhoads RE, et al. Enteroviral protease 2A cleaves dystrophin:

evidence of cytoskeletal disruption in an acquired cardiomyopathy.

Nat Med 1999;5:320–6.

[44] Seko Y, Shinkai Y, Kawasaki A, Yagita H, Okumura K, Yazaki Y.

Evidence of perforin-mediated cardiac myocyte injury in acute

murine myocarditis caused by coxsackie virus B3. J Pathol 1993;

170:53–8.

[45] Godeny EK, Gauntt CJ. Involvement of natural killer cells in

coxsackievirus B3 viral-induced myocarditis. J Immunol 1986;137:

1695–702.

[46] Godeny EK, Gauntt CJ. Murine natural killer cells limit coxsackie

virus B3 replication. J Immunol 1987;139:913–8.

[47] Seko Y, Takahashi N, Yagita H, Okumura K, Yazaki Y. Expression of

cytokine mRNAs in murine hearts with acute myocarditis caused by

Coxsackievirus B3. J Pathol 1997;183:105–8.

[48] Mason JW, O’Connell JB, Herskowitz A, Rose NR, McManus BM,

Billingham ME, et al. A clinical trial of immunosuppressive therapy

for myocarditis. The myocarditis treatment trial investigators. N Engl

J Med 1995;333:269–75.

[49] McCarthy RE 3rd, Boehmer JP, Hruban RH, Hutchins GM, Kasper

EK, Hare JM, et al. Long-term outcome of fulminant myocarditis as

compared with (nonfulminat) myocarditis. N Engl J Med 2000;85:

499–504.

[50] Herzum M, Weller R, Jomaa H, Wietrzychowski F, Pankuweit S,

Mahr P, et al. Left ventricular hemodynamic parameters in the course

of acute experimental coxsackievirus B 3 myocarditis. J Mol Cell

Cardiol 1995;27:1573–80.

[51] Kyu B, Matsumori A, Sato Y, Okada I, Chapman NM, Tracy S.

Cardiac persistence of cardioviral RNA detected by polymerase

chain reaction in a murine model of dilated cardiomyopathy.

Circulation 1992;86:522–30.

[52] Klingel K, Hohenadl C, Canu A, Albrecht M, Seemann M, Mall G, et

al. Ongoing enterovirus-induced myocarditis is associated with

persistent heart muscle infection: quantitative analysis of virus

replication, tissue damage, and inflammation. Proc Natl Acad Sci

U S A 1992;89:314–8.

[53] Lenzo JC, Fairweather D, Cull V, Shellam GR, Lawson CM.

Characterisation of murine cytomegalovirus myocarditis: cellular

infiltration of the heart and virus persistence. J Mol Cell Cardiol

2002;34:629–40.

[54] Wessely R, Klingel K, Santana LF, Dalton N, Hongo M, Jonathan

Lederer W, et al. Transgenic expression of replication-restricted

enteroviral genomes in heart muscle induces defective excitation-

contraction coupling and dilated cardiomyopathy. J Clin Invest

1998;102:1444–53.

[55] Fakhouri F, Placier S, Ardaillou R, Dussaule JC, Chatziantoniou C.

Angiotensin II activates collagen type I gene in the renal cortex and

aorta of transgenic mice through interaction with endothelin and

TGF-h. J Am Soc Nephrol 2001;12:2701–10.

[56] Smits JFM, van Krimpen C, Schoemaker RG, Cleutjens JP, Daemen

MJ. Angiotensin II receptor blockade after myocardial infarction in

rats: effects on hemodynamics, myocardial DNA synthesis, and

interstitial collagen content. J Cardiovasc Pharmacol 1992;20:772–8.

[57] Li YY, McTiernan CF, Feldman AM. Proinflammatory cytokines

regulate tissue inhibitors of metalloproteinases and disintegrin

metalloproteinase in cardiac cells. Cardiovasc Res 1999;42:162–72.

[58] Weber KT, Anversa P, Armstrong PW, Brilla CG, Burnett JC Jr,

Cruickshank JM, et al. Remodeling and reparation of the cardiovas-

cular system. J Am Coll Cardiol 1992;30:3–16.

[59] Woessner JF Jr, Nagase H. Introduction to the matrix metal-

loproteinases (MMPs). Matrix Metalloproteinases and TIMPs. New

York’ Oxford University Press; 2000. p. 1–10.

[60] Woessner JF Jr. The matrix metalloproteinase family. In: Parks WC,

Mecham RP, editors. Matrix Metalloproteinases. San Diego, Calif’

Academic Press; 1998. p. 1–14.

[61] Jeffrey JJ. Interstitial collagenases. In: Parks WC, Mecham P,

editors. Matrix Metalloproteinases. San Diego’ Academic Press;

1998. p. 15–42.

[62] Coker ML, Thomas CV, Clari MJ, Hendrick JW, Krombach RS,

Galis ZS, et al. Myocardial matrix metalloproteinase activity and

abundance with congestive heart failure. Am J Physiol 1998;274:

H1516–23.

[63] Coker ML, Doscher MA, Thomas CV, Galis ZS, Spinale FG. Matrix

metalloproteinase activity and expression in isolated LV myocyte

preparations. Am J Physiol 1999;277:H777–87.

[64] Nagaoka I, Hirota S. Increased expression of matrix metalloprotei-

nase-9 in neutrophils in glycogen-induced peritoneal inflammation of

guinea pigs. Inflamm Res 2000;49:55–62.

[65] Tao ZY, Cavasin MA, Yang F, Liu YH, Yang XP. Temporal changes

in matrix metalloproteinase expression and inflammatory response

associated with cardiac rupture after myocardial infarction in mice.

Life Sci 2004;74:1561–72.

[66] Miyamori H, Takino T, Seiki M, Sato H. Human membrane type-

2 matrix metalloproteinase is defective in cell-associated activation

of progelatinase A. Biochem Biophys Res Commun 2000;267:

796–800.

[67] Maquart FX, Pickart L, Laurent M, Gillery P, Monboisse JC, Borel

JP. Stimulation of collagen synthesis in fibroblast cultures by the

tripeptide-copper complex glycyl-l-histidyl-l-lysine-Cu2+. FEBS

Lett 1988;238:343–6.

[68] Diekmann O, Tschesche H. Degradation of kinins, angiotensins and

substance P by polymorphonuclear matrix metalloproteinases MMP

8 and MMP 9. Braz J Med Biol Res 1994;27:1865–76.

[69] Fowlkes JL, Enghild JJ, Suzuki K, Nagase H. Matrix metal-

loproteinases degrade insulin-like growth factor-binding protein-3

in dermal fibroblast cultures. J Biol Chem 1994;269:25742–6.

[70] Kridel SJ, Chen E, Kotra LP, Howard EW, Mobashery S, Smith JW.

Substrate hydrolysis by matrix metalloproteinase-9. J Biol Chem

2001;276:20572–8.

[71] Egeblad M, Werb Z. New functions for the matrix metalloproteinases

in cancer progression. Nat Rev Cancer 2002;2:163–76.

[72] Emonard H, Grimaud JA. Matrix metalloproteinases. A review. Cell

Mol Biol 1990;36:131–53.

[73] Benbow U, Brinckerhoff CE. The AP-1 site and MMP gene

regulation: What is all the fuss about? Matrix Biol 1997;15:519–26.

[74] Bond M, Chase AJ, Baker AH, Newby AC. Inhibition of

transcription factor NF-nB reduces matrix metalloproteinase-1, -3

and -9 production by vascular smooth muscle cells. Cardiovasc Res

2001;50:556–65.

[75] Eberhardt W, Huwiler A, Beck KF, Walpen S, Pfeilschifter J.

Amplification of IL-1h-induced matrix metalloproteinase-9 expres-

sion by superoxide in rat glomerular mesangial cells is mediated by

S. Rutschow et al. / Cardiovascular Research 69 (2006) 646–656 655

by guest on June 1, 2013http://cardiovascres.oxfordjournals.org/

Dow

nloaded from

increased activities of matrix metalloproteinase-9 and activating

protein-1 and involves activation of the mitogen-activated protein

kinase pathways. J Immunol 2000;165:5788–97.

[76] Hozumi A, Nishimura Y, Nishiuma T, Kotani Y, Yokoyama M.

Induction of MMP-9 in normal human bronchial epithelial cells by

TNFa via NF-nB-mediated pathway. Am J Physiol, Lung Cell Mol

Physiol 2001;281:L1444–52.

[77] Futamura M, Kamiya S, Tsukamoto M, Hirano A, Monden Y,

Arakawa H, et al. A potent inhibitor of transcription controlled by the

Ras responsive element, inhibits Rasmediated transformation activity

with suppression of MMP-1 and MMP-9 in NIH3T3 cells. Oncogene

2001;20:6724–30.

[78] Thant AA, Nawa A, Kikkawa F, Ichigotani Y, Zhang Y, Sein TT, et

al. Fibronectin activates matrix metalloproteinase-9 secretion via the

MEK1-MAPK and the PI3K-AKT pathways in ovarian cancer cells.

Clin Exp Metastasis 2001;18:423–8.

[79] Piacentini L, Gray M, Honbo NY, Chentoufi J, Bergman M,

Karliner JS. Endothelin-1 stimulates cardiac fibroblast proliferation

through activation of protein kinase C. J Mol Cell Cardiol 2000;

32:565–76.

[80] Kupfahl C, Pink D, Friedrich K, Zurbrugg HR, Neuss M, Warnecke

C, et al. Angiotensin II directly increases transforming growth factor

beta1 and osteopontin and indirectly affects collagen mRNA

expression in the human heart. Cardiovasc Res 2000;46:463–75.

[81] Zhou M, Zhang Y, Ardans JA, Wahl LM. Interferon-g differentially

regulates monocyte matrix metalloproteinase-1 and-9 through tumor

necrosis factor-a and caspase 8. J Biol Chem 2003;46:45406–13.

[82] Wahl SM, Allen JB,Weeks BS,Wong HL, Klotman PE. Transforming

growth factor h enhances integrin expression and type IV collagenase

secretion in human monocytes. Proc Natl Acad Sci U S A

1993;90:4577–81.

[83] Coker ML, Jolly JR, Joffs C, Etoh T, Holder JR, Bond BR, et al.

Matrix metalloproteinase expression and activity in isolated myo-

cytes after neurohormonal stimulation. Am J Physiol, Heart Circ

Physiol 2001;281:H543–51.

[84] Reinhardt D, Sigusch HH, Hensse J, Tyagi SC, Korfer R, Figulla HR.

Cardiac remodeling 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.

[85] Matsutomo K, Makino N, Fushiki MS. Effects of losartan on the

collagen degradative enzymes in hypertrophic and congestive types

of cardiomyopathic hamsters. Mol Cell Biochem 2001;224:19–27.

[86] Rosano L, Varmi M, Salani D, Di Castro V, Spinella F, Natali PG, et

al. Endothelin-1 induces tumor proteinase activation and invasive-

ness of ovarian carcinoma cells. Cancer Res 2001;61:8340–6.

[87] Tsuruda T, Boerrigter G, Huntley BK, Noser JA, Cataliotti A,

Costello-Boerrigter LC, et al. Brain natriuretic peptide is produced in

cardiac fibroblasts and induces matrix metalloproteinases. Circ Res

2002;91:1127–34.

[88] He CS, Wilhelm SM, Pentland AP, Marmer BL, Grant GA, Eisen

AZ, et al. Tissue cooperation in a proteolytic cascade activating

human interstitial collagenase. Proc Natl Acad Sci USA 1989;86:

2632–6.

[89] Heymans S, Lupu F, Terclavers S, Vanwetswinkel B, Herbert JM,

Baker A, et al. Loss or inhibition of uPA or MMP-9 attenuates LV

remodeling and dysfunction after acute pressure overload in mice.

Am J Pathol 2005;166:15–25.

[90] Saksela O, Rifkin DB. Cell-associated plasminogen activation:

regulation and physiological functions. Annu Rev Cell Biol 1988;4:

93–126.

[91] Nerlov C, Rorth P, Blasi F, Johnson M. Essential AP-1 and PEA3

binding elements in the human urokinase enhancer display cell type-

specific activity. Oncogene 1991;6:1583–92.

[92] Ogura N, Tobe M, Tamaki H, Nagura H, Abiko Y. Il-1h increases

uPA and uPA receptor expression in human gingival fibroblasts.

IUBMB 2001;51:381–5.

[93] Lijnen HR, Arza B, Van Hoef B, Collen D, Declerck PJ. Inactivation

of plasminogen activator inhibitor-1 by specific proteolysis with

stromelysin-1 (MMP-3). J Biol Chem 2000;275:37645–50.

[94] Lijnen HR, Van Hoef B, Collen D. Inactivation of the serpin a (2)-

antiplasmin by stromelysin-1. Biochim Biophys Acta 2001;1547:

206–13.

[95] Spinale FG. Matrix metalloproteinases: regulation and dysregulation

in the failing heart. Circ Res 2002;90:520–30.

[96] Li YY, McTiernan CF, Feldman AM. Interplay of matrix metal-

loproteinases, tissue inhibitors of metalloproteinases and their

regulators in cardiac matrix remodeling. Cardiovasc Res 2000;46:

214–24.

[97] Ries C, Petrides PE. Cytokine regulation of matrix metalloproteinase

activity and its regulatory dysfunction in disease. Biol Chem 1995;

376:345–55.

[98] Goldberg GI, Marmer BL, Grant GA, Eisen AZ, Wilhelm S, He C.

Human 72-kilodalton type IV collagenase forms a complex with a

tissue inhibitor of metalloprotease designated TIMP. Proc Natl Acad

Sci Am 1989;86:8207–11.

[99] Zucker S, Lysik RM, Zarrabi HM, Moll U, Tickle SP, Stetler-

Stevenson W, et al. Plasma assay of matrix metalloproteinases

(MMPs) and MMPinhibitor complexes in cancer. Potential use in

predicting metastasis and monitoring treatment. Ann NY Acad Sci

1994;732:248–62.

[100] Amour A, Slocombe PM, Webster A, Butler M, Knight CG, Smith

BJ, et al. TNF-alpha converting enzyme (TACE) is inhibited by

TIMP-3. FEBS Lett 1998;435:39–44.

[101] Brew A, Dinakarpandian D, Nagase H. Tissue inhibitors of metal-

loproteinases: evolution, structure and function1. Biochim Biophys

Acta 2000;1477:267–83.

[102] Nagase H, Itoh Y, Binner S. Interaction of a2-Macroglobulin with

matrix metalloproteinases and its use for identification of their active

forms. Annals Natl Acad Sci 1994;732:294–302.

[103] Moutsiakis D, Mancuso P, Krutzsch H, Stetler-Stevenson W, Zucker

S. Characterization of metalloproteinases and tissue inhibitors of

metallopteinases in human plasma. Connect Tissue Res 1992;28:

213–30.

[104] Woodiwiss AJ, Tsotetsi OJ, Sprott S, Lancaster EJ, Mela T, Chung

ES, et al. Reduction in myocardial collagen cross-linking parallels

left ventricular dilatation in rat models of systolic chamber

dysfunction. Circulation 2001;103:155–60.

[105] Okada Y, Naka K, Kawamura K, Matsumoto T, Nakanishi I,

Fujimoto N, et al. Localization of matrix metalloproteinase 9 (92

kilodalton gelatinase/type IV collagenase=gelatinase B) in osteo-

clasts: implications for bone resorption. Lab Invest 1995;72:311–22.

[106] 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 experimen-

tal myocardial infarction. J Clin Invest 2000;106:55–62.

[107] Pauschinger M, Hoppe K, Schwimmbeck PL. Relevance of myocar-

dial inflammation for the activation of matrix metalloproteinase

expression in patients with inflammatory cardiomyopathy. Circula-

tion 2003;108:IV–24.

[108] Schwartzkopff B, Fassbach M, Pelzer B, Brehm M, Strauer BE.

Elevated serum markers of collagen degradation in patients with

mild to moderate dilated cardiomyopathy. Eur J Heart Fail 2002;

4:439–44.

[109] McCormick RJ, Thomas D. Collagen crosslinking in the heart:

relationship to development and function. Basic Appl Myol 1998;

8:143–50.

[110] Gunja Smith Z, Morales AR, Romanelli R, Woessner Jr JF.

Remodelling of human myocardial collagen in idiopathic dilated

hypertrophy. Am J Pathol 1996;148:1639–48.

[111] Klappacher G, Franzen P, Haab D, Mehrabi M, Binder M, Plesch K,

et al. Measuring extracellular matrix turnover in the serum of patients

with idiopathic or ischemic dilated cardiomyopathy and impact on

dignosis and prognosis. Am J Cardiol 1995;75:913–8.

S. Rutschow et al. / Cardiovascular Research 69 (2006) 646–656656

[112] Kuhl U, Pauschinger M, Schwimmbeck PL, Seeberg B, Lober C,

Noutsias M, et al. Interferon- h treatment eliminates cardiotropic

viruses and improves left ventricular function in patients with

myocardial persistence of viral genomes and left ventricular

dysfunction. Circulation 2003;107:2793–8.

[113] Tschope C, Westermann D, Steendijk P, Noutsias M, Rutschow S,

Weitz A, et al. Hemodynamic characterization of left ventricular

function in experimental coxsackieviral myocarditis: effects of

carvedilol and metoprolol. Eur J Pharmacol 2004;491:173–9.

[114] Nishio R, Shioi T, Sasayama S, Matsumori A. Carvedilol increases

the production of interleukin-12 and interferon-g and improves the

survival of mice infected with the encephalomyocarditis virus. JACC

2003;41:340–5.

[115] Lopez BL, Christopher TA, Yue TL, Ruffolo R, Fenerstein GZ, Ma

XL. Carvedilol, a new h-adrenoreceptor blocker antihypertensive

drug, protects against free-radical induced endothelial dysfunction.

Pharmacology 1995;51:165–73.

[116] Yue TL, McKenna PJ, Gu JL, Cheng HY, Ruffolo Jr RR, Feuerstein

GZ. Carvedilol, a new vasodilating adrenoceptor blocker antihyper-

tensive drug, protects endothelial cells from damage initiated by

xanthine–xanthine oxidase and neutrophils. Cardiovasc Res 1994;

28:400–6.

[117] Yue TL, McKenna PJ, Lysko PG, Ruffolo Jr RR, Feuerstein GZ.

Carvedilol, a new antihypertensive, prevents oxidation of human low

density lipoprotein by macrophages and copper. Atherosclerosis

1992;97:209–13.

by guest on June 1, 2013http://cardiovascres.oxfordjournals.org/

Dow

nloaded from