Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Toxicol Sci ; 159(1): 189-202, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28903498

ABSTRACT

Bisphenol A (BPA) leaches from plastics to contaminate foodstuffs. Analogs, such as bisphenol S (BPS), are now used increasingly in manufacturing. Greater BPA exposure has been correlated with exacerbation of cardiovascular disease, including myocardial infarction (MI). To test the hypothesis that bisphenol exposure impairs cardiac healing, we exposed C57bl/6n mice to water containing 25ng/ml BPA or BPS from conception and surgically induced an MI in adult male progeny. Increased early death and cardiac dilation, and reduced cardiac function were found post-MI in BPA- and BPS-exposed mice. Flow cytometry revealed increased monocyte and macrophage infiltration that correlated with increased chemokine C-C motif ligand-2 expression in the infarct. In vitro BPA and BPS addition increased matrix metalloproteinase-9 (MMP) protein and secreted activity in RAW264.7 macrophage cells suggesting that invivo increases in MMP2 and MMP9 in exposed infarcts were myeloid-derived. Bone marrow-derived monocytes isolated from exposed mice had greater expression of pro-inflammatory polarization markers when chemokine stimulated indicating an enhanced susceptibility to develop a pro-inflammatory monocyte population. Chronic BPA exposure of estrogen receptor beta (ERß) deficient mice did not worsen early death, cardiac structure/function, or expression of myeloid markers after an MI. In contrast, BPS exposure of ERß-deficient mice resulted in greater death and expression of myeloid markers. We conclude that lifelong exposure to BPA or BPS augmented the monocyte/macrophage inflammatory response and adverse remodeling from an MI thereby reducing the ability to survive and successfully recover, and that the adverse effect of BPA, but not BPS, is downstream of ERß signaling.


Subject(s)
Benzhydryl Compounds/toxicity , Myocardial Infarction/physiopathology , Phenols/toxicity , Sulfones/toxicity , Animals , Estrogen Receptor beta/genetics , Heart Function Tests , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Myocardial Infarction/enzymology , Myocardial Infarction/metabolism , RAW 264.7 Cells
2.
Toxicol Sci ; 146(1): 101-15, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25862758

ABSTRACT

Estrogenic compounds such as bisphenol A (BPA) leach from plastics into food and beverage containers. Increased BPA exposure has been correlated with increased cardiovascular disease. To test the hypothesis that increased BPA exposure reduces cardiovascular remodeling, we chronically exposed C57bl/6n male mice to BPA and performed a myocardial infarction (MI). We measured cardiac function, as well as myeloid and cardiac fibroblast accumulation and activity. We found increased early death as well as increased cardiac dilation and reduced cardiac function in surviving BPA-exposed mice. Matrix metalloproteinase-2 (MMP2) protein and activity were increased 1.5-fold in BPA-exposed heart. BPA-exposed mice had similar neutrophil infiltration; however, monocyte and macrophage (MΦ) infiltration into the ischemic area was 5-fold greater than VEH mice potentially due to a 2-fold increase in monocyte chemoattractant protein-1. Monocyte and MΦ exposure to BPA in vitro in primary bone marrow cultures or in isolated peritoneal MΦ increased polarization to an activated MΦ, increased MMP2 and MMP9 expression 2-fold and activity 3-fold, and increased uptake of microspheres 3-fold. Cardiac fibroblasts (CF) differentiate to α-smooth muscle actin (αSMA) expressing myofibroblasts, migrate to the ischemic area and secrete collagen to strengthen the scar. Collagen and αSMA expression were reduced 50% in BPA-exposed hearts. Chronic in vivo or continuous in vitro BPA exposure ablated transforming growth factor beta-mediated differentiation of CF, reduced αSMA expression 50% and reduced migration 40% yet increased secreted MMP2 activity 2-fold. We conclude that chronic BPA exposure reduces the ability to successfully remodel after an MI by increasing MΦ-based inflammation and reducing myofibroblast repair function.


Subject(s)
Benzhydryl Compounds/toxicity , Heart/drug effects , Myocardial Infarction/physiopathology , Phenols/toxicity , Animals , Benzhydryl Compounds/administration & dosage , Heart Function Tests , Male , Mice , Mice, Inbred C57BL , Phenols/administration & dosage
3.
Can J Physiol Pharmacol ; 91(9): 741-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23984849

ABSTRACT

The detrimental effects of in utero exposure to the non-steroidal estrogen diethylstilbestrol (DES) are particularly marked in women. Fetal hearts express estrogen receptors, making them potentially responsive to DES. To examine whether gestational exposure to DES would impact the heart, we exposed pregnant C57bl/6n dams to DES (0.1, 1.0, and 10.0 µg·(kg body mass)(-1)·day(-1)) on gestation days 11.5-14.5, and examined the measured cardiac structure/function and calcium homeostasis protein expression in adult females. At baseline, echocardiography revealed eccentric hypertrophy in mice treated with 10.0 µg·(kg body mass)(-1)·day(-1) DES, and immunoblots showed increased SERCA2a in all DES-treated mice. Mice were swim-trained to assess cardiac remodeling. Swim-trained vehicle-treated mice developed eccentric hypertrophy without changing SERCA2 or calsequestrin 2 expression. In contrast, no DES-treated mice hypertrophied, and all increased in SERCA2a and calsequestrin 2 expression after training. To determine whether DES-induced changes in DNA methylation is part of the mechanism for its long-term effects, we measured DNA methyltransferase expression and DNA methylation. Global DNA methylation and DNA methyltransferase 3a expression were unchanged. However, DES-treated mice had increased DNA methylation in the calsequestrin 2 promoter. Thus, gestational exposure to DES altered female ventricular DNA, cardiac structure/function, and calcium homeostasis protein expression. We conclude that gestational exposure to estrogenizing compounds may impact cardiac structure/function in adult females.


Subject(s)
DNA Methylation/drug effects , Diethylstilbestrol/toxicity , Fetal Heart/drug effects , Heart Ventricles/drug effects , Hypertrophy, Left Ventricular/chemically induced , Prenatal Exposure Delayed Effects , Ventricular Function, Left/drug effects , Age Factors , Animals , Base Sequence , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Calsequestrin/genetics , Calsequestrin/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Female , Fetal Heart/growth & development , Fetal Heart/metabolism , Gene Expression Regulation , Gestational Age , Heart Ventricles/diagnostic imaging , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Hemodynamics/drug effects , Hypertrophy, Left Ventricular/diagnostic imaging , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Pregnancy , Promoter Regions, Genetic , RNA, Messenger/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sedentary Behavior , Sex Factors , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/metabolism , Swimming , Ultrasonography , Ventricular Remodeling/drug effects
4.
Toxicol Appl Pharmacol ; 266(1): 27-37, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23142472

ABSTRACT

Pregnant women, and thus their fetuses, are exposed to many endocrine disruptor compounds (EDCs). Fetal cardiomyocytes express sex hormone receptors making them potentially susceptible to re-programming by estrogenizing EDCs. Diethylstilbestrol (DES) is a proto-typical, non-steroidal estrogen. We hypothesized that changes in adult cardiac structure/function after gestational exposure to the test compound DES would be a proof in principle for the possibility of estrogenizing environmental EDCs to also alter the fetal heart. Vehicle (peanut oil) or DES (0.1, 1.0 and 10.0µg/kg/da.) was orally delivered to pregnant C57bl/6n dams on gestation days 11.5-14.5. At 3months, male progeny were left sedentary or were swim trained for 4weeks. Echocardiography of isoflurane anesthetized mice revealed similar cardiac structure/function in all sedentary mice, but evidence of systolic dysfunction and increased diastolic relaxation after swim training at higher DES doses. The calcium homeostasis proteins, SERCA2a, phospholamban, phospho-serine 16 phospholamban and calsequestrin 2, are important for cardiac contraction and relaxation. Immunoblot analyses of ventricle homogenates showed increased expression of SERCA2a and calsequestrin 2 in DES mice and greater molecular remodeling of these proteins and phospho-serine 16 phospholamban in swim trained DES mice. DES increased cardiac DNA methyltransferase 3a expression and DNA methylation in the CpG island within the calsequestrin 2 promoter in heart. Thus, gestational DES epigenetically altered ventricular DNA, altered cardiac function and expression, and reduced the ability of adult progeny to cardiac remodel when physically challenged. We conclude that gestational exposure to estrogenizing EDCs may impact cardiac structure/function in adult males.


Subject(s)
DNA Methylation/physiology , Diethylstilbestrol/toxicity , Estrogens, Non-Steroidal/toxicity , Heart/growth & development , Prenatal Exposure Delayed Effects/chemically induced , Protein Biosynthesis/physiology , Age Factors , Animals , DNA Methylation/drug effects , DNA Methyltransferase 3A , Female , Heart/drug effects , Male , Mice , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/physiopathology , Protein Biosynthesis/drug effects , Structure-Activity Relationship
5.
Am J Physiol Heart Circ Physiol ; 301(4): H1706-15, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21803942

ABSTRACT

Calcium flux into and out of the sarco(endo)plasmic reticulum is vitally important to cardiac function because the cycle of calcium entry and exit controls contraction and relaxation. Putative estrogen and androgen consensus binding sites near to a CpG island are present in the cardiac calsequestrin 2 (CSQ2) promoter. Cardiomyocytes express sex hormone receptors and respond to sex hormones. We hypothesized that sex hormones control CSQ2 expression in cardiomyocytes and so affect cardiac structure/function. Echocardiographic analysis of male and female C57bl6n mice identified thinner walled and lighter hearts in females and significant concentric remodeling after long-term gonadectomy. CSQ2 and sodium-calcium exchanger-1 (NCX1) expression was significantly increased in female compared with male hearts and decreased postovariectomy. NCX1, but not CSQ2, expression was increased postcastration. CSQ2 expression was reduced when H9c2 cells were cultured in hormone-deficient media; increased when estrogen receptor-α (ERα), estrogen receptor-ß (ERß), or androgen agonists were added; and increased in hearts from ERß-deficient mice. CSQ2 expression was reduced in mice fed a diet low in the methyl donor folic acid and in cells treated with 5-azadeoxycytidine suggesting an involvement of DNA methylation. DNA methylation in CpG in the CSQ2 CpG island was significantly different in males and females and was additionally changed postgonadectomy. Expression of DNA methyltransferases 1, 3a, and 3b was unchanged. These studies strongly link sex hormone-directed changes in CSQ2 expression to DNA methylation with changed expression correlated with altered left ventricular structure and function.


Subject(s)
Gonadal Steroid Hormones/physiology , Ventricular Function, Left/physiology , Animals , Base Sequence , Calcium/physiology , Calsequestrin/genetics , Calsequestrin/physiology , Cell Line , DNA Methylation , Echocardiography , Estrogen Receptor beta/genetics , Estrogen Receptor beta/physiology , Female , Gene Expression Regulation , Gonadal Steroid Hormones/biosynthesis , Gonadal Steroid Hormones/genetics , Homeostasis/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Myocardium/cytology , Orchiectomy , Ovariectomy , Sodium-Calcium Exchanger/genetics
6.
Vascul Pharmacol ; 51(5-6): 350-8, 2009.
Article in English | MEDLINE | ID: mdl-19815094

ABSTRACT

Therapy with bisphosphonates, including alendronate (ALN), is considered a safe and effective treatment for osteoporosis. However, recent studies have reported an unexpected increase in serious atrial fibrillation (AF) in patients treated with bisphosphonates. The mechanism that explains this side effect remains unknown. Since AF is associated with an altered sarcoendoplasmic reticulum calcium load, we studied how ALN affects cardiomyocyte calcium homeostasis and protein isoprenylation in vitro. Acute and long-term (48h) treatment of atrial and ventricular cardiomyocytes with ALN (10(-8)-10(-6)M) was performed. Changes in calcium dynamics were determined by both fluorescence measurement of cytosolic free Ca(2+) concentration and western blot analysis of calcium-regulating proteins. Finally, effect of ALN on protein farnesylation was also identified. In both atrial and ventricular cardiomyocytes, ALN treatment delayed and diminished calcium responses to caffeine. Only in atrial cells, long-term exposure to ALN-induced transitory calcium oscillations and led to the development of oscillatory component in calcium responses to caffeine. Changes in calcium dynamics were accompanied by changes in expression of proteins controlling sarcoendoplasmic reticulum calcium. In contrast, ALN minimally affected protein isoprenylation in these cells. In summary, treatment of atrial cardiomyocytes with ALN-induced abnormalities in calcium dynamics consistent with induction of a self-stimulatory, pacemaker-like behavior, which may contribute to the development of cardiac side effects associated with these drugs.


Subject(s)
Alendronate/pharmacology , Bone Density Conservation Agents/pharmacology , Calcium/metabolism , Myocytes, Cardiac/drug effects , Animals , Caffeine/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Myocytes, Cardiac/metabolism , Protein Prenylation , Sarcoplasmic Reticulum Calcium-Transporting ATPases/analysis
7.
Cardiovasc Res ; 81(4): 695-702, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19103607

ABSTRACT

AIMS: The transcription factor early growth response-1 (Egr-1) is increased in models of cardiac pathology; however, it is unclear how Egr-1 impacts the heart. We sought to identify how Egr-1 regulates expression of proteins involved in cardiomyocyte calcium homeostasis. METHODS: Protein expression was measured by immunoblotting in control cardiac differentiated H9c2 cells or in H9c2 cells overexpressing wild-type Egr-1 (Egr-1) or an Egr-1 (I293F) mutant. Microspectrofluorimetry of fura-2-loaded cells was used to study calcium dynamics. Chromatin immunoprecipitation with anti-Egr-1 antibody was used to identify Egr-1-associated DNA. RESULTS: Calsequestrin (CSQ) expression was reduced in Egr-1- and profoundly reduced in I293F-expressing cells. Calreticulin, triadin, sarcoendoplasmic reticulum ATPase 2a, phospholamban, and phosphoserine 16-phospholamban expression was unaffected. Calcium release from CSQ-dependent ryanodine-sensitive stores was reduced in Egr-1 and absent in I293F-expressing cells. In contrast, calcium release from calreticulin-dependent inositol 1,4,5-trisphosphate stores was unaffected. In vivo and in vitro chromatin immunoprecipitation demonstrated Egr-1 binding to the CSQ2 promoter. The Egr-1-binding region contains overlapping Egr-1, SP1, and nuclear factor of activated T-cells (NFAT) sites and a CpG island. Reciprocal immunoprecipitation coupled to immunoblots indicated Egr-1:NFAT3 binding was present in all cells lines. Treatment with cyclosporin A, inhibition of DNA methylation using 5-azadeoxycytidine, or inhibition of protein acetylation using sodium butyrate reduced CSQ expression. CONCLUSION: Our data suggest that Egr-1:DNA binding at the promoter, DNA methylation, and protein acetylation are important in CSQ repression. Moreover, we demonstrate that a reduction in CSQ protein is associated with abnormal calcium dynamics. We conclude that Egr-1 acts as a transcriptional repressor at the CSQ promoter, resulting in downregulation of CSQ, the major calcium storage protein that links excitation-contraction coupling in the cardiac sarcoendoplasmic reticulum.


Subject(s)
Calcium Signaling , Calsequestrin/metabolism , Early Growth Response Protein 1/metabolism , Myocytes, Cardiac/metabolism , Acetylation , Animals , Binding Sites , Calcium-Binding Proteins , Calsequestrin/genetics , Carrier Proteins/metabolism , Cell Line , DNA Methylation , Down-Regulation , Early Growth Response Protein 1/genetics , Endoplasmic Reticulum/metabolism , Heart Ventricles/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mutation , NFATC Transcription Factors/metabolism , Promoter Regions, Genetic , Protein Processing, Post-Translational , Rats , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...