Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Toxicol Sci ; 185(1): 64-76, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34718823

ABSTRACT

Benzene is a ubiquitous environmental pollutant abundant in household products, petrochemicals, and cigarette smoke. Benzene is a well-known carcinogen in humans and experimental animals; however, little is known about the cardiovascular toxicity of benzene. Recent population-based studies indicate that benzene exposure is associated with an increased risk for heart failure. Nonetheless, it is unclear whether benzene exposure is sufficient to induce and/or exacerbate heart failure. We examined the effects of benzene (50 ppm, 6 h/day, 5 days/week, and 6 weeks) or high-efficiency particulate absorbing-filtered air exposure on transverse aortic constriction (TAC)-induced pressure overload in male C57BL/6J mice. Our data show that benzene exposure had no effect on cardiac function in the Sham group; however, it significantly compromised cardiac function as depicted by a significant decrease in fractional shortening and ejection fraction, as compared with TAC/Air-exposed mice. RNA-seq analysis of the cardiac tissue from the TAC/benzene-exposed mice showed a significant increase in several genes associated with adhesion molecules, cell-cell adhesion, inflammation, and stress response. In particular, neutrophils were implicated in our unbiased analyses. Indeed, immunofluorescence studies showed that TAC/benzene exposure promotes infiltration of CD11b+/S100A8+/myeloperoxidase+-positive neutrophils in the hearts by 3-fold. In vitro, the benzene metabolites, hydroquinone, and catechol, induced the expression of P-selectin in cardiac microvascular endothelial cells by 5-fold and increased the adhesion of neutrophils to these endothelial cells by 1.5- to 2.0-fold. Benzene metabolite-induced adhesion of neutrophils to the endothelial cells was attenuated by anti-P-selectin antibody. Together, these data suggest that benzene exacerbates heart failure by promoting endothelial activation and neutrophil recruitment.


Subject(s)
Heart Failure , Ventricular Remodeling , Animals , Benzene/toxicity , Endothelial Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Ventricular Remodeling/physiology
2.
Am J Physiol Heart Circ Physiol ; 321(4): H784-H797, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34533403

ABSTRACT

Coenzyme A (CoA) is an essential cofactor required for intermediary metabolism. Perturbations in homeostasis of CoA have been implicated in various pathologies; however, whether CoA homeostasis is changed and the extent to which CoA levels contribute to ventricular function and remodeling during pressure overload has not been explored. In this study, we sought to assess changes in CoA biosynthetic pathway during pressure overload and determine the impact of limiting CoA on cardiac function. We limited cardiac CoA levels by deleting the rate-limiting enzyme in CoA biosynthesis, pantothenate kinase 1 (Pank1). We found that constitutive, cardiomyocyte-specific Pank1 deletion (cmPank1-/-) significantly reduced PANK1 mRNA, PANK1 protein, and CoA levels compared with Pank1-sufficient littermates (cmPank1+/+) but exerted no obvious deleterious impact on the mice at baseline. We then subjected both groups of mice to pressure overload-induced heart failure. Interestingly, there was more ventricular dilation in cmPank1-/- during the pressure overload. To explore potential mechanisms contributing to this phenotype, we performed transcriptomic profiling, which suggested a role for Pank1 in regulating fibrotic and metabolic processes during the pressure overload. Indeed, Pank1 deletion exacerbated cardiac fibrosis following pressure overload. Because we were interested in the possibility of early metabolic impacts in response to pressure overload, we performed untargeted metabolomics, which indicated significant changes to metabolites involved in fatty acid and ketone metabolism, among other pathways. Collectively, our study underscores the role of elevated CoA levels in supporting fatty acid and ketone body oxidation, which may be more important than CoA-driven, enzyme-independent acetylation in the failing heart.NEW & NOTEWORTHY Changes in CoA homeostasis have been implicated in a variety of metabolic diseases; however, the extent to which changes in CoA homeostasis impacts remodeling has not been explored. We show that limiting cardiac CoA levels via PANK deletion exacerbated ventricular remodeling during pressure overload. Our results suggest that metabolic alterations, rather than structural alterations, associated with Pank1 deletion may underlie the exacerbated cardiac phenotype during pressure overload.


Subject(s)
Energy Metabolism , Myocardium/enzymology , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Ventricular Dysfunction, Left/enzymology , Ventricular Function, Left , Ventricular Remodeling , Animals , Aorta/physiopathology , Aorta/surgery , Apoptosis , Arterial Pressure , Coenzyme A/metabolism , Disease Models, Animal , Female , Fibrosis , Gene Deletion , Humans , Male , Metabolome , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Transcriptome , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology
3.
PLoS One ; 15(11): e0242250, 2020.
Article in English | MEDLINE | ID: mdl-33253217

ABSTRACT

RATIONALE: The beta-O-linkage of N-acetylglucosamine (i.e., O-GlcNAc) to proteins is a pro-adaptive response to cellular insults. To this end, increased protein O-GlcNAcylation improves short-term survival of cardiomyocytes subjected to acute injury. This observation has been repeated by multiple groups and in multiple models; however, whether increased protein O-GlcNAcylation plays a beneficial role in more chronic settings remains an open question. OBJECTIVE: Here, we queried whether increasing levels of cardiac protein O-GlcNAcylation would be beneficial during infarct-induced heart failure. METHODS AND RESULTS: To achieve increased protein O-GlcNAcylation, we targeted Oga, the gene responsible for removing O-GlcNAc from proteins. Here, we generated mice with cardiomyocyte-restricted, tamoxifen-inducible haploinsufficient Oga gene. In the absence of infarction, we observed a slight reduction in ejection fraction in Oga deficient mice. Overall, Oga reduction had no major impact on ventricular function. In additional cohorts, mice of both sexes and both genotypes were subjected to infarct-induced heart failure and followed for up to four weeks, during which time cardiac function was assessed via echocardiography. Contrary to our prediction, the Oga deficient mice exhibited exacerbated-not improved-cardiac function at one week following infarction. When the observation was extended to 4 wk post-MI, this acute exacerbation was lost. CONCLUSIONS: The present findings, coupled with our previous work, suggest that altering the ability of cardiomyocytes to either add or remove O-GlcNAc modifications to proteins exacerbates early infarct-induced heart failure. We speculate that more nuanced approaches to regulating O-GlcNAcylation are needed to understand its role-and, in particular, the possibility of cycling, in the pathophysiology of the failing heart.


Subject(s)
Myocardial Infarction/pathology , Myocardium/enzymology , N-Acetylglucosaminyltransferases/genetics , Ventricular Dysfunction/etiology , Animals , Echocardiography , Female , Glycosylation , Haploinsufficiency , Heart/physiology , Heart Failure/metabolism , Heart Failure/pathology , Humans , Male , Mice , Mice, Knockout , Middle Aged , Myocardial Infarction/complications , Myocardium/metabolism , Myocardium/pathology , N-Acetylglucosaminyltransferases/deficiency , N-Acetylglucosaminyltransferases/metabolism , Tamoxifen/pharmacology , Up-Regulation , Ventricular Function/drug effects
4.
Basic Res Cardiol ; 114(4): 28, 2019 05 31.
Article in English | MEDLINE | ID: mdl-31152247

ABSTRACT

Several post-translational modifications figure prominently in ventricular remodeling. The beta-O-linkage of N-acetylglucosamine (O-GlcNAc) to proteins has emerged as an important signal in the cardiovascular system. Although there are limited insights about the regulation of the biosynthetic pathway that gives rise to the O-GlcNAc post-translational modification, much remains to be elucidated regarding the enzymes, such as O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), which regulate the presence/absence of O-GlcNAcylation. Recently, we showed that the transcription factor, E2F1, could negatively regulate OGT and OGA expression in vitro. The present study sought to determine whether E2f1 deletion would improve post-infarct ventricular function by de-repressing expression of OGT and OGA. Male and female mice were subjected to non-reperfused myocardial infarction (MI) and followed for 1 or 4 week. MI significantly increased E2F1 expression. Deletion of E2f1 alone was not sufficient to alter OGT or OGA expression in a naïve setting. Cardiac dysfunction was significantly attenuated at 1-week post-MI in E2f1-ablated mice. During chronic heart failure, E2f1 deletion also attenuated cardiac dysfunction. Despite the improvement in function, OGT and OGA expression was not normalized and protein O-GlcNAcyltion was not changed at 1-week post-MI. OGA expression was significantly upregulated at 4-week post-MI but overall protein O-GlcNAcylation was not changed. As an alternative explanation, we also performed guided transcriptional profiling of predicted targets of E2F1, which indicated potential differences in cardiac metabolism, angiogenesis, and apoptosis. E2f1 ablation increased heart size and preserved remote zone capillary density at 1-week post-MI. During chronic heart failure, cardiomyocytes in the remote zone of E2f1-deleted hearts were larger than wildtype. These data indicate that, overall, E2f1 exerts a deleterious effect on ventricular remodeling. Thus, E2f1 deletion improves ventricular remodeling with limited impact on enzymes regulating O-GlcNAcylation.


Subject(s)
E2F1 Transcription Factor/deficiency , Myocardial Infarction/metabolism , Myocardium/metabolism , Ventricular Function, Left , Ventricular Remodeling , Animals , Capillaries/metabolism , Capillaries/pathology , Coronary Vessels/metabolism , Coronary Vessels/pathology , Disease Models, Animal , E2F1 Transcription Factor/genetics , Female , Gene Deletion , Glycosylation , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , N-Acetylglucosaminyltransferases/metabolism , beta-N-Acetylhexosaminidases/metabolism
5.
Redox Biol ; 17: 440-449, 2018 07.
Article in English | MEDLINE | ID: mdl-29885625

ABSTRACT

Pathological cardiac remodeling during heart failure is associated with higher levels of lipid peroxidation products and lower abundance of several aldehyde detoxification enzymes, including aldehyde dehydrogenase 2 (ALDH2). An emerging idea that could explain these findings concerns the role of electrophilic species in redox signaling, which may be important for adaptive responses to stress or injury. The purpose of this study was to determine whether genetically increasing ALDH2 activity affects pressure overload-induced cardiac dysfunction. Mice subjected to transverse aortic constriction (TAC) for 12 weeks developed myocardial hypertrophy and cardiac dysfunction, which were associated with diminished ALDH2 expression and activity. Cardiac-specific expression of the human ALDH2 gene in mice augmented myocardial ALDH2 activity but did not improve cardiac function in response to pressure overload. After 12 weeks of TAC, ALDH2 transgenic mice had larger hearts than their wild-type littermates and lower capillary density. These findings show that overexpression of ALDH2 augments the hypertrophic response to pressure overload and imply that downregulation of ALDH2 may be an adaptive response to certain forms of cardiac pathology.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/genetics , Heart Failure/genetics , Oxidative Stress/genetics , Ventricular Remodeling/genetics , Animals , Aorta/metabolism , Gene Expression Regulation , Heart Failure/metabolism , Heart Failure/pathology , Humans , Mice , Mice, Transgenic , Mitochondria/metabolism , Myocardium/metabolism , Myocardium/pathology , Oxidation-Reduction , Pressure , Signal Transduction/genetics
6.
J Mol Cell Cardiol ; 118: 183-192, 2018 05.
Article in English | MEDLINE | ID: mdl-29627295

ABSTRACT

Pathological cardiac hypertrophy is associated with the accumulation of lipid peroxidation-derived aldehydes such as 4-hydroxy-trans-2-nonenal (HNE) and acrolein in the heart. These aldehydes are metabolized via several pathways, of which aldose reductase (AR) represents a broad-specificity route for their elimination. We tested the hypothesis that by preventing aldehyde removal, AR deficiency accentuates the pathological effects of transverse aortic constriction (TAC). We found that the levels of AR in the heart were increased in mice subjected to TAC for 2 weeks. In comparison with wild-type (WT), AR-null mice showed lower ejection fraction, which was exacerbated 2 weeks after TAC. Levels of atrial natriuretic peptide and myosin heavy chain were higher in AR-null than in WT TAC hearts. Deficiency of AR decreased urinary levels of the acrolein metabolite, 3-hydroxypropylmercapturic acid. Deletion of AR did not affect the levels of the other aldehyde-metabolizing enzyme - aldehyde dehydrogenase 2 in the heart, or its urinary product - (N-Acetyl-S-(2-carboxyethyl)-l-cystiene). AR-null hearts subjected to TAC showed increased accumulation of HNE- and acrolein-modified proteins, as well as increased AMPK phosphorylation and autophagy. Superfusion with HNE led to a greater increase in p62, LC3II formation, and GFP-LC3-II punctae formation in AR-null than WT cardiac myocytes. Pharmacological inactivation of JNK decreased HNE-induced autophagy in AR-null cardiac myocytes. Collectively, these results suggest that during hypertrophy the accumulation of lipid peroxidation derived aldehydes promotes pathological remodeling via excessive autophagy, and that metabolic detoxification of these aldehydes by AR may be essential for maintaining cardiac function during early stages of pressure overload.


Subject(s)
Aldehyde Reductase/deficiency , Autophagy , Heart/physiopathology , Pressure , Aldehyde Reductase/metabolism , Aldehydes/metabolism , Animals , Aorta/pathology , Cardiomegaly/diagnostic imaging , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Constriction, Pathologic , Gene Deletion , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice, Inbred C57BL , Myocardial Contraction , Myocardium/enzymology , Sequestosome-1 Protein/metabolism
7.
Circ Res ; 122(10): 1347-1353, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29483092

ABSTRACT

RATIONALE: Increasing evidence indicates the presence of lncRNAs in various cell types. Airn is an imprinting gene transcribed from the paternal chromosome. It is in antisense orientation to the imprinted, but maternally derived, Igf2r gene, on which Airn exerts its regulation in cis. Although Airn is highly expressed in the heart, functions aside from imprinting remain unknown. OBJECTIVE: Here, we studied the functions of Airn in the heart, especially cardiomyocytes. METHODS AND RESULTS: Silencing of Airn via siRNAs augmented cell death, vulnerability to cellular stress, and reduced cell migration. To find the cause of such phenotypes, the potential binding partners of Airn were identified via RNA pull-down followed by mass spectrometry, which indicated Igf2bp2 (insulin-like growth factor 2 mRNA-binding protein 2) and Rpa1 (replication protein A1) as potential binding partners. Further experiments showed that Airn binds to Igf2bp2 to control the translation of several genes. Moreover, silencing of Airn caused less binding of Igf2bp2 to other mRNAs and reduced translation of Igf2bp2 protein. CONCLUSIONS: Our study uncovers a new function of Airn and demonstrates that Airn is important for the physiology of cardiomyocytes.


Subject(s)
Myocytes, Cardiac/metabolism , RNA, Long Noncoding/genetics , RNA-Binding Proteins/biosynthesis , Animals , Cell Line , Cell Movement , Gene Expression Regulation , Mice , Myocardial Infarction/metabolism , Organ Specificity , Protein Binding , Protein Biosynthesis , RNA Interference , RNA Splicing , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , RNA-Binding Proteins/genetics , Replication Protein A/metabolism
8.
Basic Res Cardiol ; 112(3): 23, 2017 05.
Article in English | MEDLINE | ID: mdl-28299467

ABSTRACT

The myocardial response to pressure overload involves coordination of multiple transcriptional, posttranscriptional, and metabolic cues. The previous studies show that one such metabolic cue, O-GlcNAc, is elevated in the pressure-overloaded heart, and the increase in O-GlcNAcylation is required for cardiomyocyte hypertrophy in vitro. Yet, it is not clear whether and how O-GlcNAcylation participates in the hypertrophic response in vivo. Here, we addressed this question using patient samples and a preclinical model of heart failure. Protein O-GlcNAcylation levels were increased in myocardial tissue from heart failure patients compared with normal patients. To test the role of OGT in the heart, we subjected cardiomyocyte-specific, inducibly deficient Ogt (i-cmOgt -/-) mice and Ogt competent littermate wild-type (WT) mice to transverse aortic constriction. Deletion of cardiomyocyte Ogt significantly decreased O-GlcNAcylation and exacerbated ventricular dysfunction, without producing widespread changes in metabolic transcripts. Although some changes in hypertrophic and fibrotic signaling were noted, there were no histological differences in hypertrophy or fibrosis. We next determined whether significant differences were present in i-cmOgt -/- cardiomyocytes from surgically naïve mice. Interestingly, markers of cardiomyocyte dedifferentiation were elevated in Ogt-deficient cardiomyocytes. Although no significant differences in cardiac dysfunction were apparent after recombination, it is possible that such changes in dedifferentiation markers could reflect a larger phenotypic shift within the Ogt-deficient cardiomyocytes. We conclude that cardiomyocyte Ogt is not required for cardiomyocyte hypertrophy in vivo; however, loss of Ogt may exert subtle phenotypic differences in cardiomyocytes that sensitize the heart to pressure overload-induced ventricular dysfunction.


Subject(s)
Cardiomegaly/metabolism , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , N-Acetylglucosaminyltransferases/metabolism , Animals , Apoptosis , Disease Models, Animal , Humans , Immunoblotting , In Situ Nick-End Labeling , Male , Mice , Mice, Knockout , Middle Aged , Polymerase Chain Reaction
9.
Circ Res ; 120(4): 692-700, 2017 Feb 17.
Article in English | MEDLINE | ID: mdl-28073804

ABSTRACT

RATIONALE: Acute kidney injury (AKI) is common during high-risk percutaneous coronary intervention (PCI), particularly in those with severely reduced left ventricular ejection fraction. The impact of partial hemodynamic support with a microaxial percutaneous left ventricular assist device (pLVAD) on renal function after high-risk PCI remains unknown. OBJECTIVE: We tested the hypothesis that partial hemodynamic support with the Impella 2.5 microaxial pLVAD during high-risk PCI protected against AKI. METHODS AND RESULTS: In this retrospective, single-center study, we analyzed data from 230 patients (115 consecutive pLVAD-supported and 115 unsupported matched-controls) undergoing high-risk PCI with ejection fraction ≤35%. The primary outcome was incidence of in-hospital AKI according to AKI network criteria. Logistic regression analysis determined the predictors of AKI. Overall, 5.2% (6) of pLVAD-supported patients versus 27.8% (32) of unsupported control patients developed AKI (P<0.001). Similarly, 0.9% (1) versus 6.1% (7) required postprocedural hemodialysis (P<0.05). Microaxial pLVAD support during high-risk PCI was independently associated with a significant reduction in AKI (adjusted odds ratio, 0.13; 95% confidence intervals, 0.09-0.31; P<0.001). Despite preexisting CKD or a lower ejection fraction, pLVAD support protection against AKI persisted (adjusted odds ratio, 0.63; 95% confidence intervals, 0.25-0.83; P=0.04 and adjusted odds ratio, 0.16; 95% confidence intervals, 0.12-0.28; P<0.001, respectively). CONCLUSIONS: Impella 2.5 (pLVAD) support protected against AKI during high-risk PCI. This renal protective effect persisted despite the presence of underlying CKD and decreasing ejection fraction.


Subject(s)
Acute Kidney Injury/prevention & control , Heart-Assist Devices/trends , Hemodynamics/physiology , Percutaneous Coronary Intervention/trends , Postoperative Complications/prevention & control , Acute Kidney Injury/etiology , Aged , Female , Heart-Assist Devices/adverse effects , Humans , Male , Middle Aged , Percutaneous Coronary Intervention/adverse effects , Postoperative Complications/etiology , Retrospective Studies , Risk Factors , Treatment Outcome
10.
Front Cell Dev Biol ; 4: 78, 2016.
Article in English | MEDLINE | ID: mdl-27536657

ABSTRACT

Cell therapy improves cardiac function. Few cells have been investigated more extensively or consistently shown to be more effective than c-kit sorted cells; however, c-kit expression is easily lost during passage. Here, our primary goal was to develop an improved method to isolate c-kit(pos) cells and maintain c-kit expression after passaging. Cardiac mesenchymal cells (CMCs) from wild-type mice were selected by polystyrene adherence properties. CMCs adhering within the first hours are referred to as rapidly adherent (RA); CMCs adhering subsequently are dubbed slowly adherent (SA). Both RA and SA CMCs were c-kit sorted. SA CMCs maintained significantly higher c-kit expression than RA cells; SA CMCs also had higher expression endothelial markers. We subsequently tested the relative efficacy of SA vs. RA CMCs in the setting of post-infarct adoptive transfer. Two days after coronary occlusion, vehicle, RA CMCs, or SA CMCs were delivered percutaneously with echocardiographic guidance. SA CMCs, but not RA CMCs, significantly improved cardiac function compared to vehicle treatment. Although the mechanism remains to be elucidated, the more pronounced endothelial phenotype of the SA CMCs coupled with the finding of increased vascular density suggest a potential pro-vasculogenic action. This new method of isolating CMCs better preserves c-kit expression during passage. SA CMCs, but not RA CMCs, were effective in reducing cardiac dysfunction. Although c-kit expression was maintained, it is unclear whether maintenance of c-kit expression per se was responsible for improved function, or whether the differential adherence property itself confers a reparative phenotype independently of c-kit.

11.
J Biol Chem ; 290(52): 31013-24, 2015 Dec 25.
Article in English | MEDLINE | ID: mdl-26527687

ABSTRACT

Protein O-GlcNAcylation, which is controlled by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), has emerged as an important posttranslational modification that may factor in multiple diseases. Until recently, it was assumed that OGT/OGA protein expression was relatively constant. Several groups, including ours, have shown that OGT and/or OGA expression changes in several pathologic contexts, yet the cis and trans elements that regulate the expression of these enzymes remain essentially unexplored. Here, we used a reporter-based assay to analyze minimal promoters and leveraged in silico modeling to nominate several candidate transcription factor binding sites in both Ogt (i.e. the gene for OGT protein) and Mgea5 (i.e. the gene for OGA protein). We noted multiple E2F binding site consensus sequences in both promoters. We performed chromatin immunoprecipitation in both human and mouse cells and found that E2F1 bound to candidate E2F binding sites in both promoters. In HEK293 cells, we overexpressed E2F1, which significantly reduced OGT and MGEA5 expression. Conversely, E2F1-deficient mouse fibroblasts had increased Ogt and Mgea5 expression. Of the known binding partners for E2F1, we queried whether retinoblastoma 1 (Rb1) might be involved. Rb1-deficient mouse embryonic fibroblasts showed increased levels of Ogt and Mgea5 expression, yet overexpression of E2F1 in the Rb1-deficient cells did not alter Ogt and Mgea5 expression, suggesting that Rb1 is required for E2F1-mediated suppression. In conclusion, this work identifies and validates some of the promoter elements for mouse Ogt and Mgea5 genes. Specifically, E2F1 negatively regulates both Ogt and Mgea5 expression in an Rb1 protein-dependent manner.


Subject(s)
Antigens, Neoplasm/biosynthesis , E2F1 Transcription Factor/metabolism , Gene Expression Regulation, Enzymologic/physiology , Histone Acetyltransferases/biosynthesis , Hyaluronoglucosaminidase/biosynthesis , N-Acetylglucosaminyltransferases/biosynthesis , Response Elements/physiology , 3T3-L1 Cells , Animals , Antigens, Neoplasm/genetics , E2F1 Transcription Factor/genetics , HEK293 Cells , Histone Acetyltransferases/genetics , Humans , Hyaluronoglucosaminidase/genetics , Mice , Mice, Mutant Strains , N-Acetylglucosaminyltransferases/genetics , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism
13.
Biochem J ; 467(1): 115-26, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25627821

ABSTRACT

Diabetes is characterized by hyperglycaemia and perturbations in intermediary metabolism. In particular, diabetes can augment flux through accessory pathways of glucose metabolism, such as the hexosamine biosynthetic pathway (HBP), which produces the sugar donor for the ß-O-linked-N-acetylglucosamine (O-GlcNAc) post-translational modification of proteins. Diabetes also promotes mitochondrial dysfunction. Nevertheless, the relationships among diabetes, hyperglycaemia, mitochondrial dysfunction and O-GlcNAc modifications remain unclear. In the present study, we tested whether high-glucose-induced increases in O-GlcNAc modifications directly regulate mitochondrial function in isolated cardiomyocytes. Augmentation of O-GlcNAcylation with high glucose (33 mM) was associated with diminished basal and maximal cardiomyocyte respiration, a decreased mitochondrial reserve capacity and lower Complex II-dependent respiration (P<0.05); however, pharmacological or genetic modulation of O-GlcNAc modifications under normal or high glucose conditions showed few significant effects on mitochondrial respiration, suggesting that O-GlcNAc does not play a major role in regulating cardiomyocyte mitochondrial function. Furthermore, an osmotic control recapitulated high-glucose-induced changes to mitochondrial metabolism (P<0.05) without increasing O-GlcNAcylation. Thus, increased O-GlcNAcylation is neither sufficient nor necessary for high-glucose-induced suppression of mitochondrial metabolism in isolated cardiomyocytes.


Subject(s)
Acetylglucosamine/metabolism , Down-Regulation , Glucose/metabolism , Hyperglycemia/metabolism , Mitochondria, Heart/metabolism , Oxidative Phosphorylation , Protein Processing, Post-Translational , Aminoacylation , Animals , Animals, Newborn , Cells, Cultured , Electron Transport Complex II/antagonists & inhibitors , Electron Transport Complex II/metabolism , Energy Metabolism , Glucose/adverse effects , Mitochondria, Heart/enzymology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Osmolar Concentration , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Up-Regulation , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/metabolism
14.
J Biol Chem ; 289(43): 29665-76, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25183011

ABSTRACT

Derangements in metabolism and related signaling pathways characterize the failing heart. One such signal, O-linked ß-N-acetylglucosamine (O-GlcNAc), is an essential post-translational modification regulated by two enzymes, O-GlcNAc transferase and O-GlcNAcase (OGA), which modulate the function of many nuclear and cytoplasmic proteins. We recently reported reduced OGA expression in the failing heart, which is consistent with the pro-adaptive role of increased O-GlcNAcylation during heart failure; however, molecular mechanisms regulating these enzymes during heart failure remain unknown. Using miRNA microarray analysis, we observed acute and chronic changes in expression of several miRNAs. Here, we focused on miR-539 because it was predicted to target OGA mRNA. Indeed, co-transfection of the OGA-3'UTR containing reporter plasmid and miR-539 overexpression plasmid significantly reduced reporter activity. Overexpression of miR-539 in neonatal rat cardiomyocytes significantly suppressed OGA expression and consequently increased O-GlcNAcylation; conversely, the miR-539 inhibitor rescued OGA protein expression and restored O-GlcNAcylation. In conclusion, this work identifies the first target of miR-539 in the heart and the first miRNA that regulates OGA. Manipulation of miR-539 may represent a novel therapeutic target in the treatment of heart failure and other metabolic diseases.


Subject(s)
Heart Failure/genetics , MicroRNAs/metabolism , Up-Regulation/genetics , beta-N-Acetylhexosaminidases/metabolism , 3' Untranslated Regions/genetics , Animals , Base Sequence , Cell Hypoxia/genetics , Down-Regulation/genetics , Glycosylation , HEK293 Cells , Heart Function Tests , Humans , Mice, Inbred C57BL , MicroRNAs/genetics , Molecular Sequence Data , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Oxygen/metabolism , Rats, Sprague-Dawley
15.
Pharmacol Ther ; 142(1): 62-71, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24287310

ABSTRACT

The cardiovascular system is capable of robust changes in response to physiologic and pathologic stimuli through intricate signaling mechanisms. The area of metabolism has witnessed a veritable renaissance in the cardiovascular system. In particular, the post-translational ß-O-linkage of N-acetylglucosamine (O-GlcNAc) to cellular proteins represents one such signaling pathway that has been implicated in the pathophysiology of cardiovascular disease. This highly dynamic protein modification may induce functional changes in proteins and regulate key cellular processes including translation, transcription, and cell death. In addition, its potential interplay with phosphorylation provides an additional layer of complexity to post-translational regulation. The hexosamine biosynthetic pathway generally requires glucose to form the nucleotide sugar, UDP-GlcNAc. Accordingly, O-GlcNAcylation may be altered in response to nutrient availability and cellular stress. Recent literature supports O-GlcNAcylation as an autoprotective response in models of acute stress (hypoxia, ischemia, oxidative stress). Models of sustained stress, such as pressure overload hypertrophy, and infarct-induced heart failure, may also require protein O-GlcNAcylation as a partial compensatory mechanism. Yet, in models of Type II diabetes, O-GlcNAcylation has been implicated in the subsequent development of vascular, and even cardiac, dysfunction. This review will address this apparent paradox and discuss the potential mechanisms of O-GlcNAc-mediated cardioprotection and cardiovascular dysfunction. This discussion will also address potential targets for pharmacologic interventions and the unique considerations related to such targets.


Subject(s)
Acetylglucosamine/metabolism , Cardiovascular System/metabolism , Animals , Exercise , Hexosamines/metabolism , Humans , Protein Processing, Post-Translational
16.
J Thorac Cardiovasc Surg ; 147(5): 1634-43, 2014 May.
Article in English | MEDLINE | ID: mdl-24139617

ABSTRACT

OBJECTIVE: Left ventricular assist device support produces a bleeding diathesis. Evidence suggests a major role for von Willebrand factor (vWF). We examined vWF metabolism in a preclinical model of short-term mechanical circulatory support. METHODS: In 25 calves (weight, 80-110 kg), the inflow/outflow graft of the Symphony Heart Assist System was sewn end-to-side to the carotid artery. Support was initiated (acute, n = 4; 1 week, n = 16; 2 weeks, n = 5). Acutely, carotid artery pressure and flow were measured to evaluate the hemodynamic changes near the anastomosis. At baseline and after ≤2 weeks of support, platelet aggregometry with adenosine 5'-diphosphate, collagen, and ristocetin was performed. Gel electrophoresis and wet immunoblotting qualitatively evaluated vWF multimers and quantified plasma ADAMTS-13, the vWF-cleaving protease. Carotid arterial rings near the anastomosis were studied with immunohistochemical staining for ADAMTS-13 and were cultured to quantify endothelial ADAMTS-13 production. Fluorescent resonance energy transfer was used to evaluate the enzymatic activity of ADAMTS-13 in the plasma and in supernatant from cultured carotid arterial rings. Plasma interleukin-6, which inhibits ADAMTS-13 activity, was measured using an enzyme-linked immunosorbent assay. RESULTS: During support, statistically significant (P < .05) changes in the carotid endothelium arterial hemodynamics were observed. The highest molecular weight vWF multimers were absent, and the vWF-ristocetin platelet aggregation pathway was significantly impaired. A modest but significant increase in plasma ADAMTS-13 protein and activity was observed. ADAMTS-13 decreased significantly in the carotid near the anastomosis but increased significantly in supernatant from cultured carotid arterial rings. The plasma interleukin-6 levels did not change significantly. CONCLUSIONS: Hemodynamic activation of vWF and increased plasma ADAMTS-13 activity may have reduced high-molecular-weight vWF multimers and thereby impaired the vWF-platelet aggregation pathway. Additional delineation of these pathways may improve management of left ventricular assist device-associated bleeding.


Subject(s)
Carotid Arteries/surgery , Heart-Assist Devices , Platelet Aggregation , von Willebrand Factor/metabolism , ADAM Proteins/blood , Animals , Carotid Arteries/metabolism , Cattle , Endothelial Cells/metabolism , Heart-Assist Devices/adverse effects , Hemodynamics , Hemorrhage/blood , Hemorrhage/etiology , Interleukin-6/blood , Male , Models, Animal , Molecular Weight , Platelet Function Tests , Prosthesis Design , Time Factors , Tissue Culture Techniques , Ventricular Function, Left
17.
ASAIO J ; 59(2): 123-9, 2013.
Article in English | MEDLINE | ID: mdl-23438773

ABSTRACT

In patients with a ventricular assist device (VAD), diminished high-molecular-weight von Willebrand factor (vWF) multimers may contribute to a bleeding diathesis. The mechanistic pathway(s) of vWF degradation and the role of ADAMTS-13, the vWF-cleaving metalloproteinase, are unknown. The objective of this study was to investigate the molecular mechanisms of VAD-induced vWF impairment in an in vitro system.Simple, mock circulatory loops (n = 4) were developed with a clinically approved, paracorporeal continuous-flow VAD. The loops were primed with anticoagulated, whole bovine blood (750 ml). The VAD was operated at constant blood flow and pressure. Blood samples were drawn at baseline and hourly for 6 hours. vWF multimers and ADAMTS-13 protein were quantified by agarose and polyacrylamide gel electrophoresis with immunoblotting. Plasma platelet factor 4 (PF4), a marker of platelet activation, was quantified via ELISA.Within 120 minutes, high-molecular-weight vWF multimers decreased, and low-molecular-weight multimers increased. Multiple low-molecular-weight vWF fragments emerged (~140, 176, 225, and 310 kDa). Total plasma ADAMTS-13 increased by 13 ± 3% (p < 0.05). Plasma PF4 increased by 21 ± 7% (p = 0.05).During VAD support, vWF degradation occurred quickly. Multiple mechanisms were responsible and included vWF cleavage by ADAMTS-13 (140 and 176 kDa fragments), and what may have been mechanical demolition of endogenous plasma vWF (225 kDa fragments) and nascent vWF (225 and 310 kDa fragments) from platelets. A modest increase in plasma ADAMTS-13 from activated platelets may have contributed to this process but was not the major mechanism. Mechanical demolition was likely the dominant process and warrants further evaluation.


Subject(s)
Heart-Assist Devices , von Willebrand Diseases/blood , von Willebrand Factor/metabolism , ADAM Proteins/blood , ADAM Proteins/physiology , ADAMTS13 Protein , Animals , Cattle , Molecular Weight , Platelet Factor 4/blood , Shear Strength , von Willebrand Diseases/surgery
18.
J Surg Res ; 172(1): e55-60, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22079842

ABSTRACT

BACKGROUND: The Matrigel assay provides a versatile platform to examine vessel growth. Similarly, the microsphere method is used extensively in laboratory animals to measure tissue-specific blood flow. However, microsphere models have not been used with Matrigel to study angiogenesis in live animals. The goal of this study was to develop a novel technique to directly measure blood flow with microspheres in vessels grown in Matrigel in vivo. METHODS: In calves (n = 10, 110 ± 5 kg), 5 mL of Matrigel was injected subcutaneously. After 10 d, a percutaneous cardiac catheterization was performed. Fluorescent-labeled 15 µm microspheres were injected into the left ventricular chamber to distribute throughout the body based on systemic blood flow patterns. Afterwards, Matrigel plugs were removed, and animals were recovered. Flow cytometry was used to count microspheres and quantify blood flow within the plug. FITC-conjugated isolectin-B4 staining was performed to quantify Matrigel capillary density. Flow cytometry was performed to quantify circulating plasma CD34(+) cells. Linear regressions were used to determine relationships between Matrigel blood flow, Matrigel capillary density, and plasma CD34(+) cells. RESULTS: Over 10 d, small-caliber vessels grew into subcutaneous Matrigel plugs. Microspheres lodged throughout the plug and indicated that newly grown vessels in the Matrigel were functional and able to accommodate blood flow. Modest associations between Matrigel blood flow, Matrigel capillary density, and circulating plasma CD34(+) cells were noted. CONCLUSION: This method provides a novel and cost-effective technique to measure blood flow within vessels grown in Matrigel in vivo.


Subject(s)
Collagen , Laminin , Microspheres , Microvessels/physiology , Neovascularization, Physiologic/physiology , Proteoglycans , Regional Blood Flow/physiology , Animals , Antigens, CD34/metabolism , Capillaries/physiology , Cattle , Drug Combinations , Flow Cytometry , Male , Plasma Cells/cytology , Plasma Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...