Modulation of Cardiac Mast Cell Mediated Extracellular Matrix ...

8 downloads 0 Views 187KB Size Report
Feb 18, 2005 - Jason D. Gardner. David B. Murray .... Protein concentration of LV homogenates was determined using BCA reagent (Pierce, Rockford, IL).
Articles in PresS. Am J Physiol Heart Circ Physiol (February 18, 2005). doi:10.1152/ajpheart.00765.2004

Modulation of Cardiac Mast Cell Mediated Extracellular Matrix Degradation by Estrogen

Amanda L. Chancey Jason D. Gardner David B. Murray Gregory L. Brower Joseph S. Janicki

Department of Anatomy, Physiology, and Pharmacology Auburn University, Auburn, AL 36849

Running Head: H-00765-2004.R1; Estrogen and Cardiac Mast Cells

Address Correspondence To:

Joseph S. Janicki, Ph.D. Dept. of Anatomy, Physiology, and Pharmacology 106 Greene Hall Auburn University Auburn, AL 36849-5517 (334) 844-3700 (office) (334) 844-3697 (fax) [email protected] (email)

Copyright © 2005 by the American Physiological Society.

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 2

Abstract There are fundamental differences between males and females with regard to susceptibility to heart disease. Although numerous animal models of heart failure have demonstrated that premenopausal females are afforded cardioprotection and therefore fare better in the face of cardiac disease than their male counterparts, many questions as to how this occurs still exist. Recently, we have shown: 1) that increased mast cell density is associated with adverse ventricular remodeling; and 2) chemically induced mast cell degranulation using compound 48/80 resulted in remarkable changes in matrix metalloproteinase (MMP) activity, cardiac collagen structure, and cardiac diastolic function in normal male rats. With the known gender differences in cardiac disease in mind, we sought to examine the effects of chemically induced cardiac mast cell degranulation in isolated, blood-perfused hearts of intact females, ovariectomized females, and ovariectomized females treated with 17$-estradiol. In response to mast cell degranulation, no significant differences in cardiac function, MMP-2 activity, or collagen volume fraction (CVF) were observed between intact females or ovariectomized females treated with estrogen. Whereas, in the ovariectomized female group, a significant rightward shift in the left ventricular pressure-volume relationship was noted post 48/80, accompanied by a marked 133 % increase in active MMP-2 values over that seen in intact female hearts post 48/80 (p < 0.05) and a significant reduction in CVF below control (0.46 + 0.23 vs. 0.73 + 0.13, respectively; p < 0.05). These findings indicate that estrogen’s cardioprotective role can be partially mediated by its effects on cardiac mast cells, MMPs, and the extracellular matrix. Keywords: isolated heart, matrix metalloproteinase, gender, ventricular remodeling

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 3

Introduction Despite the fact that gender differences in the prevalence and severity of cardiovascular disease have been identified in humans and in animal models of heart failure (24; 36), the mechanisms mediating cardioprotection in premenopausal females are poorly understood. We recently reported that male rats respond to chronic biventricular volume overload induced by an aortocaval (AV) fistula with a predictable pattern of significant left ventricular (LV) hypertrophy and dilatation and accompanying cardiac dysfunction (8). In contrast, female rats subjected to chronic volume overload did not develop LV dilatation and maintained compensated cardiac function with appropriate myocardial hypertrophy (14). Mast cell mediated activation of matrix metalloproteinases (MMPs) is one mechanism contributing to the development of ventricular dilatation in males. This finding was established by previous studies demonstrating an association between increased mast cell density, activation of MMPs, and the initiation of cardiac remodeling in volume overload induced by an AV fistula in male rats (4; 22) and by mitral regurgitation in dogs (38). We have also shown that mast cell degranulation induced by endothelin-1 or compound 48/80 in the isolated, blood perfused heart from male rats resulted in MMP activation, extensive collagen matrix degradation, and LV dilatation within 30 minutes (10; 31). 17$-estradiol, hereafter referred to as estrogen, has been shown to exert cardioprotective effects in animal studies (35; 37). It is interesting that a few studies have reported direct effects of estrogen on non-cardiac mast cells (16; 25), consistent with the observation that mast cells from the aorta of premenopausal women express estrogen receptors (33). Thus, the gender differences in the pattern of myocardial remodeling observed in response to an AV fistula may be related to hormonal modulation of mast cell

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 4

function. Accordingly, we sought to test the hypothesis that estrogen mediated effects on cardiac mast cells account for the lack of adverse remodeling in the female heart subjected to chronic volume overload. To this end, we sought to determine if differences in the response to acute, chemically-induced degranulation of cardiac mast cells exist among intact females, ovariectomized females, and ovariectomized females treated with estrogen. Our results indicate that the changes evoked in response to cardiac mast cell degranulation are strongly dependent on the presence of estrogen.

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 5

Materials and Methods The surgical procedures and subsequent care of the animals conformed with the principles of the National Institutes of Health “Guide for the Care and Use of Laboratory Animals,” and the protocol was approved by Auburn University’s Institutional Animal Care and Use Committee. In all terminal procedures, the animals were deeply anesthetized using sodium pentobarbital (50 mg/kg, i.p.). In order to prevent possible effects of a soy based diet, all animals were maintained on a diet that contained minimal phytoestrogens (Harlan Teklad Diet 2014).

Surgical Preparation and Experimental Protocol Age-matched adult female Sprague Dawley rats were randomly divided into groups as follows: intact females (n=6), ovariectomized females (n=7), and ovariectomized females receiving 0.02 mg/kg/day estrogen (n=6). This dose of estrogen was chosen based on preliminary data from our laboratory indicating that ovariectomized females treated with 0.02 mg/kg/day estrogen are protected from adverse myocardial remodeling induced by an AV fistula (13). Therefore, females supplemented with estrogen were implanted subcutaneously with 0.25 mg time-release pellets (Innovative Research of America, Sarasota, FL) using a 10 gauge precision trochar at the time of ovariectomy to achieve a dosage of 0.02 mg/kg/day. The ovariectomy procedure was performed two weeks prior to obtaining isolated heart function. Briefly, with the rat under halothane inhalation anesthesia, the ovaries are approached via a ventral abdominal laparotomy, the ovarian pedicle ligated and the ovary

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 6

excised. The abdominal musculature and skin were closed using standard techniques with absorbable sutures and autoclips, respectively.

Experimental Design The effects of cardiac mast cell degranulation on ventricular function was determined using an isolated, blood perfused heart preparation as previously described (8). Briefly, the ascending thoracic aorta in the anesthetized rat was cannulated for continuous retrograde perfusion of the heart via an apparatus consisting of a pressurized perfusion reservoir (100-105 mm Hg) and a venous collection reservoir connected in circuit with a female support rat. The perfusion apparatus was primed with blood obtained from female blood donors. In the case of the ovariectomized group, the support and blood donor rats were ovariectomized as well. Following positioning of a compliant balloon in the LV, pressure-volume relationships for each heart were obtained both prior to and 30 minutes after infusion of the mast cell secretagogue, compound 48/80 as previously described (10).

Administration of 1 ml of 0.9% sterile saline containing 7.2 mg of

compound 48/80 (Sigma, St. Louis, MO) to the isolated heart was accomplished by introducing the solution into the pressurized perfusion reservoir, allowing the solution to mix with the reservoir blood and perfuse through the beating heart. In a previous study, this dosage of compound 48/80 resulted in near 100% cardiac mast cell degranulation (10). The coronary venous blood containing 48/80 was not returned to the support rat. After completion of the functional studies, the atria and great vessels were removed and LV (plus septum) and right ventricle (RV) were separated and weighed. A complete transmural section of the LV at midventricle was placed in buffered formalin for fixation,

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 7

and the remainder was minced into -1 mm cubes, snap-frozen in liquid nitrogen, and stored at !80° C. The formalin-fixed tissue was processed for routine histopathology and 5 micron thick paraffin-embedded sections were stained with either pinocyanol erythrocynate for visualization of mast cell morphology or picrosirius red for quantification of collagen volume fraction (CVF), presented as the percentage of collagen area relative to total myocardial area as previously described (10). Portions of the frozen LV tissue were used to determine the wet-to-dry weight ratio as a measure of myocardial water content and for western blot analysis of protein expression. These analyses were performed with the observer blinded as to the source of the tissue sections.

Matrix Metalloproteinase Activity Matrix metalloproteinase activity in cardiac tissue extracts was performed by standard gelatin zymography procedures using a sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) matrix containing gelatin (1 mg/ml) (9).

All of the

zymograms had two lytic bands corresponding to standards for the proenzyme (68 kd) and activated (62 kd) forms of gelatinase A (MMP-2) and the proenzyme (95 kd) and activated (82 kd) forms of gelatinase B (MMP-9; Chemicon International, Inc., Temecula, CA). Each gel was run in duplicate, and in order to compare results from different gels, extract from a single heart was used as a standard on all gels. The activity of the lytic bands in the other lanes of a gel were expressed as a percent of this standard’s activity.

Once

normalized in this fashion, the percent activities from hearts belonging to each group were averaged, with the intact female group set to 100%.

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 8

Western Blot Analysis Expression of tissue inhibitor of metalloproteinase-1 (TIMP-1) and TIMP-3 protein was determined by western blotting of immunoprecipitated samples. Protein concentration of LV homogenates was determined using BCA reagent (Pierce, Rockford, IL). Equal amounts of protein (20:g) were immunoprecipitated using the protein A agarose bead separation method (Sigma, St. Louis, Mo) and monoclonal rabbit anti-human TIMP-1 or TIMP-3 antibodies (Research Diagnostic Inc, Flanders, NJ). Appropriate negative controls were performed by substituting water in place of the primary antibody prior to the addition of agarose beads. Equal amounts of supernatant (4 ul) were separated by 10% SDSPAGE and transferred onto nitrocellulose membrane (Bio-Rad Laboratories, Hercules, CA). Membranes were probed with primary antibodies for TIMP-1 and TIMP-3 (1:1000) for 4 hrs at room temperature, washed (30 min) and incubated with anti-rabbit secondary antibody conjugated to horse radish peroxidase (Amersham Biosciences Corp, Piscataway, NJ). Immunoreactive bands were visualized using enhanced chemiluminescence reagents (ECL, Amerhsam Biosciences Corp) exposed to hyperfilm at the linear range of film density. The films were scanned and densitometric analysis was performed using Un Scan It Software (Silk Scientific Inc, Orem, Utah).

Statistical Analysis Statistical analyses were performed with Systat 9.0 software (SPSS Inc., Chicago, IL). All grouped data are expressed as means ± SD, unless otherwise noted. Grouped data comparisons were made by one-way analysis of variance with intergroup comparisons analyzed using Bonferroni post-hoc testing. Statistical significance was taken to be p #

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 9

0.05. The pre- and post-48/80 pressure-volume curves for each heart were fit to a 3rd order non-linear regression (SigmaPlot, SPSS Science, Chicago, IL), and the volumes corresponding to pressure increments of 2.5 mm Hg were determined. The volumes were then averaged to obtain the final pressure-volume relationship for each group. Pressurevolume curves were analyzed using a two-factor repeated measures ANOVA.

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 10

Results Histological examination demonstrated comparable, extensive 48/80 induced mast cell degranulation in all groups (i.e., > 95%).

An increase of approximately 3% in

myocardial water also occurred in the three groups, presumably from mast cell released histamine. However, in contrast to our findings in a previous study (10), mast cell degranulation did not result in increases in MMP activity in the hearts from intact and ovariectomized + estrogen females.

It was only in the ovariectomized females not

receiving supplemental estrogen that 48/80 caused a 133% elevation in MMP-2 activity above that of intact females and 112% above that of the ovariectomized + estrogen group (p < 0.007; Figure 1). Average density of the latent MMP-2 band present was not different among groups. While bands representative of latent and active MMP-9 were present, 48/80 had no effect on the density of these bands. Moreover, the intensity of the MMP-9 bands was markedly less than that of the MMP-2 bands.

Similarly there were no

differences in TIMP-1 and TIMP-3 expression amongst the three groups (Figure 2). The significant increase in MMP-2 activity in the hearts from ovariectomized females post 48/80 was associated with a significant reduction in collagen volume fraction (CVF) below that of intact females (0.46 + 0.23 vs. 0.73 + 0.13, p < 0.05). Conversely, estrogen replacement in ovariectomized females prevented this significant 48/80 induced decrease in CVF (0.60 + 0.09, p < 0.05 vs. ovariectomized female group; Figure 3). Consistent with our previous findings of cardioprotection in intact females, hearts from normal intact females exposed to 48/80 did not develop an alteration in the LV pressure-volume curve following mast cell degranulation (Table 1). However, hearts from ovariectomized females developed a significant parallel rightward shift in the pressure-

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 11

volume relationship following exposure to 48/80. Estrogen replacement in ovariectomized females effectively prevented a 48/80 induced rightward shift in the pressure-volume relationship, reproducing the findings in intact females. Concurrent with a significant rightward shift in the pressure-volume curve post 48/80 in ovariectomized female hearts, end diastolic volume at a pressure of 0 mm Hg (V0) increased significantly (Figure 4). However, V0 was not significantly increased in the intact female and ovariectomized + estrogen female groups. Myocardial compliance, as assessed by the volume required to increase end diastolic pressure from 0 to 25 mm Hg ()V25), was not different in any of the groups post 48/80 (Table 1).

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 12

Discussion Gender differences in the susceptibility to cardiovascular disease have been established in both humans and animal models (6; 11; 14; 20; 28; 36), but the mechanisms affording this cardioprotection in females are not understood. An increased incidence of cardiovascular disease in postmenopausal women suggests that ovarian hormones may play a role in this cardioprotection (24).

Estrogen acts through receptor-mediated

pathways, and among other things has been shown to inhibit the development of atherosclerosis (2; 19), and increase endothelial and inducible nitric oxide synthase (eNOS and iNOS) activity (1; 3; 21; 34). However, reduced formation of atherosclerosis and vasodilation cannot fully account for the cardioprotective actions of estrogen. Thus, while numerous possible actions of estrogen have been examined, this is the first study to evaluate the effect of estrogen on cardiac mast cell mediated myocardial remodeling. Given our previous studies demonstrating that cardiac mast cell degranulation mediates MMP activation and extracellular matrix degradation (10; 23; 31), our hypothesis was that cardioprotection in intact females would be due to estrogen mediated prevention of mast cell degranulation. Accordingly, the purpose of this study was to determine if gender differences exist in the susceptibility to mast cell degranulation or in the initiation of myocardial remodeling following chemically-induced mast cell degranulation. However, since extensive mast cell degranulation was noted in all of the female groups stimulated with compound 48/80, mast cell stabilization does not appear to be the mechanism responsible for estrogen mediated cardioprotection. Despite our initial hypothesis being incorrect, this is the first study to demonstrate that estrogen prevents the mast cell mediated MMP activation and extracellular matrix degradation previously demonstrated in

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 13

male rats (10; 31). While this study did not address whether the estrogen cardioprotective effect was receptor mediated, preliminary data from our laboratory indicate the presence of estrogen receptors on mast cells isolated from male and female rat hearts. Therefore, greater insight into mechanisms of action will require studies using estrogen receptor antagonists. MMP-2 activity in this study was assessed using gelatinase zymography. While this technique does not directly examine collagenase activity, one would expect the activities of the various MMPs in the collagen degradation cascade to be tightly coupled (32). This was demonstrated by Gunja-Smith et al. (17), who found zymographic gelatinase activity to be closely correlated with collagenase activity determined by tritium-labeled telopeptidefree collagen degradation. Direct evidence demonstrating the ability of cardiac mast cells to activate MMPs in normal male hearts was established by recent studies from our laboratory (10). This study examined the response to chemically induced mast cell degranulation using compound 48/80, and found that cardiac mast cell degranulation resulted in a significant increase in myocardial MMP-2 activity, degradation of the myocardial extracellular matrix, and ventricular dilatation. The findings reported herein are consistent with this study in males, in that 48/80 administered to hearts from the ovariectomized female group significantly increased MMP-2 activity relative to intact females and ovariectomized + estrogen treated females. Since latent MMP-2 levels were similar for all groups, this difference in MMP-2 activity does not appear to be due to estrogen mediated suppression of MMP expression. Neither can these differences in MMP-2 activity be attributed to estrogen mediated alterations in TIMP-1 and TIMP-3 expression. Rather these findings indicate that decreased release of the mast cell

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 14

enzymes responsible for MMP activation prevented the increase in MMP activity in the intact female and ovariectomized + estrogen treated groups. In support of this conclusion is the finding of Harnish et al. (18) that non-cardiac mast cell proteases were downregulated by estrogen. The development of extracellular matrix degradation and ventricular dilatation in the ovariectomized female hearts is consistent with the observed changes in MMP-2 activity. These changes were also comparable to our previous findings demonstrating a parallel rightward shift in the pressure-volume relationship in normal male hearts post 48/80 (10). In contrast to the significant LV dilatation and degradation of interstitial collagen (i.e., CVF) in the ovariectomized group, the pressure-volume relationships prior to and following administration of 48/80 were unchanged in the hearts from the intact female and ovariectomized + estrogen rats. Estrogen replacement also effectively prevented the significant decrease in the collagen volume fraction observed in the untreated ovariectomized females. Mast cell derived histamine is known to increase vascular permeability. Accordingly, it was not surprising that myocardial edema was seen secondary to mast cell degranulation. In all likelihood this edema was responsible for the non-significant trend of increased ventricular stiffness (i.e., reduction in )V25, Table 1) post-48/80. However, the fact that the extent of myocardial edema was comparable in all three groups indicates that histamine did not contribute to the rapidly induced myocardial remodeling observed. Comparable ventricular dilatation and extracellular matrix degradation independent of histamine were reported to occur within a similar 30 minute period by MacKenna et al. (29), utilizing rat hearts perfused with bacterial collagenase.

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 15

These differences in MMP-2 activity and extracellular matrix degradation mediated by estrogen suggest an explanation for the different patterns of myocardial remodeling produced in male and female rats in the AV fistula model of heart failure (6; 8; 14). Males develop eccentric hypertrophy, marked ventricular dilatation and heart failure within 20 wks post-fistula (8), whereas intact females develop concentric hypertrophy without ventricular dilatation and ventricular function is maintained (14). However in a similar long-term study, ovariectomy eliminated this cardioprotective effect, with an AV fistula producing eccentric hypertrophy and ventricular dilatation comparable to that seen in males (6).

The

prevention of ventricular dilatation by estrogen is significant in light of the fact that ventricular dilatation is an independent risk factor in the progression to decompensated heart failure (26). Although estrogen has been shown to significantly affect MMP activity in the female reproductive system (27; 30), our understanding of the effect of estrogen on cardiac MMPs is limited (41). The study by Xu et al. (41) found that ovariectomy in otherwise normal aged rats resulted in a significant reduction in myocardial MMP-2 protein expression, with this decrease in MMP-2 expression being reversed by estrogen replacement. Conversely, while we saw no effect of ovariectomy on latent MMP-2 expression, we found a significant increase in mast cell mediated MMP-2 activation in hearts from ovariectomized rats. The reason for these disparate findings is not clear, but may be related to differences in age and duration of ovariectomy induced menopause between the studies. Estrogen has been shown to have anti-inflammatory effects (18; 25; 39; 40). Thus, alterations in mast cell content due to estrogen presents another possible mechanism. Harnish et al. (18) found that estrogen treatment was associated with a reduction in

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 16

expression of mast cell proteases in colons of diseased animals, and improved intestinal health in a rat model of inflammatory bowel disease. Further, this study demonstrated that estrogen pretreatment of bone marrow-derived mast cells repressed production of several cytokines including tumor necrosis factor-" (TNF-"), interluekin-6, and interleukin-13. While all cardiac cell types have the ability to synthesize TNF-", emerging evidence indicates that constitutive expression of TNF-" is localized in cardiac mast cells (5; 12; 15). In addition, preliminary findings indicate suppression of TNF-" synthesis and/or release by ovarian hormones as the mechanism responsible for the gender specific cardioprotection from adverse myocardial remodeling secondary to chronic volume overload (7). However, the relationship between TNF-" and MMP activation in modulating myocardial remodeling remains to be elucidated. In summary, gender differences exist in the response of the isolated heart to 48/80induced mast cell degranulation. Hearts from normal males and ovariectomized females respond to mast cell degranulation with significant increases in active MMP-2 levels, significant reductions in CVF, and a rightward shift in the pressure-volume relationship. In contrast, hearts of intact females and ovariectomized females receiving estrogen supplementation do not undergo any of these changes in response to 48/80 administration. Although current knowledge of the effects of estrogen on mast cells is limited, the results of this and other studies clearly indicate that estrogen can affect the composition and/or release of mast cell contents; which in turn affect MMP activation, extracellular matrix degradation and cardiac remodeling.

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 17

Acknowledgments This work was supported in part by NIH Grants R01-HL-62228 and R01-HL-73390 (JSJ), American Heart Association Scientist Development Grant 0435298N (JDG), American Heart Association Southern Research Consortium Postdoctoral Fellowship 0325228B (DBM) and American Heart Association Southern Research Consortium Predoctoral Fellowship 0215060B (ALC). The authors would like to thank Dr. James A. Stewart, Jr. and Mr. Steven Trott for their technical assistance.

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 18

References

1.

Andersen MR and Stender S. Endothelial nitric oxide synthase activity in aorta of

normocholesterolemic rabbits: regional variation and the effect of estrogen. Cardiovasc Res 47: 192-199, 2000. 2.

Bakir S, Mori T, Durand J, Chen YF, Thompson JA and Oparil S. Estrogen-

induced vasoprotection is estrogen receptor dependent: evidence from the balloon-injured rat carotid artery model. Circulation 101: 2342-2344, 2000. 3.

Binko J and Majewski H. 17b-Estradiol reduces vasoconstriction in endothelium-

denuded rat aortas through inducible NOS. Am J Physiol Heart Circ Physiol 274: H853H859, 1998. 4.

Brower GL, Chancey AL, Thanigaraj S, Matsubara BB and Janicki JS. Cause and

effect relationship between myocardial mast cell number and matrix metalloproteinase activity. Am J Physiol Heart Circ Physiol 283: H518-H525, 2002. 5.

Brower GL, Gardner JD, Florek JC, Holland M, Hauer-Jensen M and Janicki JS.

Cardioprotection in mast cell deficient rats with chronic volume overload. J Mol Cell Cardiol 37:214, 2004. 6.

Brower GL, Gardner JD and Janicki JS. Gender mediated cardiac protection from

adverse ventricular remodeling is abolished by ovariectomy. Mol Cell Biochem 251: 89-95, 2003. 7.

Brower GL, Gardner JD, Murray DB and Janicki JS. Gender differences in adverse

myocardial remodeling induced by tumor necrosis factor-alpha infusion in rats. Circulation

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 19

108: IV244, 2003. 8.

Brower GL, Henegar JR and Janicki JS. Temporal evaluation of left ventricular

remodeling and function in rats with chronic volume overload. Am J Physiol Heart Circ Physiol 271: H2071-H2078, 1996. 9.

Brower GL and Janicki JS. Contribution of ventricular remodeling to pathogenesis

of heart failure in rats. Am J Physiol Heart Circ Physiol 280: H674-H683, 2001. 10.

Chancey AL, Brower GL and Janicki JS. Cardiac mast cell mediated activation of

gelatinase and alteration of ventricular diastolic function. Am J Physiol Heart Circ Physiol 282: H2152-H2158, 2002. 11.

Douglas PS, Katz SE, Weinberg EO, Chen MH, Bishop SP and Lorell BH.

Hypertrophic remodeling: gender differences in the early response to left ventricular pressure overload. J Am Coll Cardiol 32: 1118-1125, 1998. 12.

Frangogiannis N, Lindsey M, Michael L, Youker K, Bressler R, Mendoza L,

Spengler R, Smith CW and Entman M. Resident cardiac mast cells degranulate and release preformed TNF-a, initiating the cytokine cascade in experimental canine myocardial ischemia/reperfusion. Circulation 98: 699-710, 1998. 13.

Gardner JD, Brower GL and Janicki JS. Cardioprotective effects of estrogen and

phytoestrogens in cardiac remodeling secondary to chronic volume overload. FASEB J 16: A1130, 2002. 14.

Gardner JD, Brower GL and Janicki JS. Gender differences in cardiac remodeling

secondary to chronic volume overload. J Card Fail 8: 101-107, 2002. 15.

Gilles S, Zahler S, Welsch U, Sommerhoff CP and Becker BF. Release of TNF-

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 20

alpha during myocardial reperfusion depends on oxidative stress and is prevented by mast cell stabilizers. Cardiovasc Res 60: 608-616, 2003. 16.

Gunin AG and Sharov AA. Role of mast cells in oestradiol effects on the uterus of

ovariectomized rats. J Reprod Fertil 113: 61-68, 1998. 17.

Gunja-Smith Z, Morales AR, Romanelli R and Woessner JF, Jr. Remodeling of

human

myocardial

collagen

in

idiopathic

dilated

cardiomyopathy.

Role

of

metalloproteinases and pyridinoline cross-links. Am J Pathol 148: 1639-1648, 1996. 18.

Harnish DC, Albert LM, Leathurby Y, Eckert AM, Ciarletta A, Kasaian M and Keith

JC, Jr. Beneficial effects of estrogen treatment in the HLA-B27 transgenic rat model of inflammatory bowel disease. Am J Physiol Gastrointest Liver Physiol 286: G118-G125, 2004. 19.

Hayashi T, Jayachandran M, Sumi D, Thakur NK, Esaki T, Muto E, Kano H, Asai

Y and Iguchi A. Physiological concentration of 17b-estradiol retards the progression of severe atherosclerosis induced by a high-cholesterol diet plus balloon catheter injury: role of NO. Arterioscler Thromb Vasc Biol 20: 1613-1621, 2000. 20.

Hayward CS, Kelly RP and Collins P. The roles of gender, the menopause and

hormone replacement on cardiovascular function. Cardiovasc Res 46: 28-49, 2000. 21.

Huang A, Sun D, Koller A and Kaley G. 17b-estradiol restores endothelial nitric

oxide release to shear stress in arterioles of male hypertensive rats. Circulation 101: 94100, 2000. 22.

Janicki JS, Brower GL, Carver W, Chancey AL, Forman MF and Jobe LJ. Role

of mast cells in cardiovascular disease. In: Cardiovascular Remodeling and Failure, edited

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 21

by Singal PK, Dixon IMC, Kirshenbaum LA and Dhalla NS. Boston: Kluwer Academic Publishers, 2003, p. 485-499. 23.

Janicki JS, Brower GL, Gardner JD, Chancey AL and Stewart JA, Jr. The dynamic

interaction between matrix metalloproteinase activity and adverse myocardial remodeling. Heart Fail Rev 9: 33-42, 2004. 24.

Kalin MF and Zumoff B. Sex hormones and coronary disease: a review of the clinical

studies. Steroids 55: 330-352, 1990. 25.

Kim MS, Chae HJ, Shin TY, Kim HM and Kim HR. Estrogen regulates cytokine

release in human mast cells. Immunopharmacol Immunotoxicol 23: 495-504, 2001. 26.

Lee TH, Hamilton MA, Stevenson LW, Moriguchi JD, Fonarow GC, Child JS,

Laks H and Walden JA. Impact of left ventricular cavity size on survival in advanced heart failure. Am J Cardiol 72: 672-676, 1993. 27.

Liotta LA. Probing the depths of degradation: matrix metalloproteinase-2 and

endometrial menstrual breakdown. J Clin Invest 97: 273-274, 1996. 28.

Litwin SE, Katz SE, Litwin CM, Morgan JP and Douglas PS. Gender differences

in postinfarction left ventricular remodeling. Cardiology 91: 173-183, 1999. 29.

MacKenna DA, Omens JH, McCulloch AD and Covell JW. Contribution of collagen

matrix to passive left ventricular mechanics in isolated rat hearts. Am J Physiol Heart Circ Physiol 266: H1007-H1018, 1994. 30.

Moalli PA, Klingensmith WL, Meyn LA and Zyczynski HM. Regulation of matrix

metalloproteinase expression by estrogen in fibroblasts that are derived from the pelvic floor. Am J Obstet Gynecol 187: 72-79, 2002.

Estrogen and Cardiac Mast Cells 31.

H-00765-2004.R1 Page 22

Murray DB, Gardner JD, Brower GL and Janicki JS. Endothelin-1 mediates cardiac

mast cell degranulation, matrix metalloproteinase activation and myocardial remodeling in rats. Am J Physiol Heart Circ Physiol 287: H2295-H2299, 2004. 32.

Nagase H. Activation mechanisms of matrix metalloproteinases. Biological Chemistry

378: 151-160, 1997. 33.

Nicovani S and Rudolph MI. Estrogen receptors in mast cells from arterial walls.

Biocell 26: 15-24, 2002. 34.

Nuedling S, Kahlert S, Loebbert K, Doevendans PA, Meyer R, Vetter H and

Grohe C. 17 Beta-estradiol stimulates expression of endothelial and inducible NO synthase in rat myocardium in-vitro and in-vivo. Cardiovasc Res 43: 666-674, 1999. 35.

Sbarouni E, Iliodromitis EK, Bofilis E, Kyriakides ZS and Kremastinos DT. Short-

term estrogen reduces myocardial infarct size in oophorectomized female rabbits in a dose-dependent manner. Cardiovasc Drugs Ther 12: 457-462, 1998. 36.

Sharkey LC, Holycross BJ, Park S, McCune SA, Hoversland R and Radin MJ.

Effect of ovariectomy in heart failure-prone SHHF/Mcc-facp rats. Am J Physiol 275: R1968R1976, 1998. 37.

Sharkey LC, Holycross BJ, Park S, Shiry LJ, Hoepf TM, McCune SA and Radin

MJ. Effect of ovariectomy and estrogen replacement on cardiovascular disease in heart failure-prone SHHF/Mcc-facp rats. J Mol Cell Cardiol 31: 1527-1537, 1999. 38.

Stewart JA, Jr, Wei CC, Brower GL, Rynders PE, Hankes GH, Dillon AR,

Lucchesi PA, Janicki JS and Dell'Italia LJ. Cardiac mast cell- and chymase-mediated matrix metalloproteinase activity and left ventricular remodeling in mitral regurgitation in the

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 23

dog. J Mol Cell Cardiol 35: 311-319, 2003. 39.

Thomas TN, Rhodin JA, Clark L, Garces A and Bryant M. A comparison of the anti-

inflammatory activities of conjugated estrogens and 17-b estradiol. Inflamm Res 52: 452460, 2003. 40.

Walsh BW, Cox DA, Sashegyi A, Dean RA, Tracy RP and Anderson PW. Role of

tumor necrosis factor-alpha and interleukin-6 in the effects of hormone replacement therapy and raloxifene on C-reactive protein in postmenopausal women. Am J Cardiol 88: 825-828, 2001. 41.

Xu Y, Arenas IA, Armstrong SJ and Davidge ST. Estrogen modulation of left

ventricular remodeling in the aged heart. Cardiovasc Res 57: 388-394, 2003.

Estrogen and Cardiac Mast Cells

H-00765-2004.R1 Page 24

Figure Legends

Figure 1.

Matrix metalloproteinase (MMP-2) activity for the three experimental groups following administration of the mast cell secretagogue compound 48/80. Data presented as mean + SEM. * p < 0.05 vs. intact female; # p < 0.05 vs. ovariectomized female treated with estrogen. OX = ovariectomized female, OX + Est = ovariectomized female treated with 0.02 mg/kg/day of estrogen.

Figure 2.

Expression of tissue inhibitor of metalloproteinase-1 and -3 (TIMP-1 and TIMP-3, respectively) protein as determined by western blotting of immunoprecipitated samples. Data presented as mean + SD.

Figure 3.

Myocardial collagen volume fraction following administration of the mast cell secretagogue compound 48/80. Data presented as mean + SD. * p < 0.05 vs. intact female; # p < 0.05 vs. ovariectomized female treated with estrogen.

Figure 4.

Percent increase in left ventricular volume at an end diastolic pressure of 0 mm Hg (V0) following administration of the mast cell secretagogue compound 48/80. Data presented as mean + SEM. * p < 0.05 vs. intact female; # p < 0.05 vs. ovariectomized female treated with estrogen.

Estrogen and Cardiac Mast Cells Table 1.

H-00765-2004.R1 Page 25

Change in V0 and )V25 in response to 48/80-induced mast cell degranulation. V0 (:l)

)V25 (:l)

pre 48/80

292.0 + 33.6

91.2 + 34.5

post 48/80

303.3 + 30.3

77.0 + 27.9

pre 48/80

312.8 + 20.3

46.1 + 16.5

post 48/80

360.2 + 21.1

36.7 + 12.3

pre 48/80

321.0 + 14.8

67.5 + 18.2

post 48/80

327.1 + 11.0

53.0 + 12.5

Group: Intact Female

OX Female

OX + Estrogen Female

Mean ± SEM.

Abbreviations:

OX = ovariectomized; V0 = left ventricular volume at an end diastolic pressure (EDP) of 0 mm Hg; )V25 = the change in LV volume between EDP 0 - 25 mm Hg.

Estrogen and Cardiac Mast Cells Figure 1.

H-00765-2004.R1 Page 26

Estrogen and Cardiac Mast Cells Figure 2.

H-00765-2004.R1 Page 27

Estrogen and Cardiac Mast Cells Figure 3.

H-00765-2004.R1 Page 28

Estrogen and Cardiac Mast Cells Figure 4.

H-00765-2004.R1 Page 29