Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 6 de 6
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Biol Chem ; 299(12): 105447, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37949223

RÉSUMÉ

The post-translational modification of intracellular proteins by O-linked ß-GlcNAc (O-GlcNAc) has emerged as a critical regulator of cardiac function. Enhanced O-GlcNAcylation activates cytoprotective pathways in cardiac models of ischemia-reperfusion (I/R) injury; however, the mechanisms underpinning O-GlcNAc cycling in response to I/R injury have not been comprehensively assessed. The cycling of O-GlcNAc is regulated by the collective efforts of two enzymes: O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), which catalyze the addition and hydrolysis of O-GlcNAc, respectively. It has previously been shown that baseline heart physiology and pathophysiology are impacted by sex. Here, we hypothesized that sex differences in molecular signaling may target protein O-GlcNAcylation both basally and in ischemic hearts. To address this question, we subjected male and female WT murine hearts to ex vivo ischemia or I/R injury. We assessed hearts for protein O-GlcNAcylation, abundance of OGT, OGA, and glutamine:fructose-6-phosphate aminotransferase (GFAT2), activity of OGT and OGA, and UDP-GlcNAc levels. Our data demonstrate elevated O-GlcNAcylation in female hearts both basally and during ischemia. We show that OGT activity was enhanced in female hearts in all treatments, suggesting a mechanism for these observations. Furthermore, we found that ischemia led to reduced O-GlcNAcylation and OGT-specific activity. Our findings provide a foundation for understanding molecular mechanisms that regulate O-GlcNAcylation in the heart and highlight the importance of sex as a significant factor when assessing key regulatory events that control O-GlcNAc cycling. These data suggest the intriguing possibility that elevated O-GlcNAcylation in females contributes to reduced ischemic susceptibility.


Sujet(s)
Acétyl-glucosamine , Coeur , Myocarde , N-acetylglucosaminyltransferase , Caractères sexuels , Transduction du signal , Animaux , Femelle , Mâle , Souris , Acétyl-glucosamine/métabolisme , Coeur/physiologie , Ischémie/enzymologie , Ischémie/métabolisme , Myocarde/enzymologie , Myocarde/métabolisme , N-acetylglucosaminyltransferase/métabolisme , Maturation post-traductionnelle des protéines
2.
Toxicol Lett ; 383: 17-32, 2023 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-37244563

RÉSUMÉ

Prenatal arsenic exposure is a major public health concern, associated with altered birth outcomes and increased respiratory disease risk. However, characterization of the long-term effects of mid-pregnancy (second trimester) arsenic exposure on multiple organ systems is scant. This study aimed to characterize the long-term impact of mid-pregnancy inorganic arsenic exposure on the lung, heart, and immune system, including infectious disease response using the C57BL/6 mouse model. Mice were exposed from gestational day 9 till birth to either 0 or 1000 µg/L sodium (meta)arsenite in drinking water. Male and female offspring assessed at adulthood (10-12 weeks of age) did not show significant effects on recovery outcomes after ischemia reperfusion injury but did exhibit increased airway hyperresponsiveness compared to controls. Flow cytometric analysis revealed significantly greater total numbers of cells in arsenic-exposed lungs, lower MHCII expression in natural killer cells, and increased percentages of dendritic cell populations. Activated interstitial (IMs) and alveolar macrophages (AMs) isolated from arsenic-exposed male mice produced significantly less IFN-γ than controls. Conversely, activated AMs from arsenic-exposed females produced significantly more IFN-γ than controls. Although systemic cytokine levels were higher upon Mycobacterium tuberculosis (Mtb) infection in prenatally arsenic-exposed offspring there was no difference in lung Mtb burden compared to controls. This study highlights significant long-term impacts of prenatal arsenic exposure on lung and immune cell function. These effects may contribute to the elevated risk of respiratory diseases associated with prenatal arsenic exposure in epidemiology studies and point to the need for more research into mechanisms driving these maintained responses.


Sujet(s)
Arsenic , Effets différés de l'exposition prénatale à des facteurs de risque , Grossesse , Souris , Mâle , Femelle , Animaux , Humains , Arsenic/toxicité , Souris de lignée C57BL , Poumon
3.
Life Sci ; 324: 121712, 2023 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-37100378

RÉSUMÉ

AIMS: Cadmium exposure is a worldwide problem that has been linked to the development of cardiovascular disease. This study aimed to elucidate mechanistic details of chronic cadmium exposure on the structure and function of the heart. MAIN METHODS: Male and female mice were exposed to cadmium chloride (CdCl2) via drinking water for eight weeks. Serial echocardiography and blood pressure measurements were performed. Markers of hypertrophy and fibrosis were assessed, along with molecular targets of Ca2+-handling. KEY FINDINGS: Males exhibited a significant reduction in left ventricular ejection fraction and fractional shortening with CdCl2 exposure, along with increased ventricular volume at end-systole, and decreased interventricular septal thickness at end-systole. Interestingly, no changes were detected in females. Experiments in isolated cardiomyocytes revealed that CdCl2-induced contractile dysfunction was also present at the cellular level, showing decreased Ca2+ transient and sarcomere shortening amplitude with CdCl2 exposure. Further mechanistic investigation uncovered a decrease in sarco/endoplasmic reticulum Ca2+-ATPase 2a (SERCA2a) protein expression and phosphorylated phospholamban levels in male hearts with CdCl2 exposure. SIGNIFICANCE: The findings of our novel study provide important insight into how cadmium exposure may act as a sex-specific driver of cardiovascular disease, and further underscore the importance of reducing human exposure to cadmium.


Sujet(s)
Maladies cardiovasculaires , Fonction ventriculaire gauche , Humains , Souris , Mâle , Femelle , Animaux , Cadmium/toxicité , Cadmium/métabolisme , Débit systolique , Maladies cardiovasculaires/métabolisme , Myocytes cardiaques/métabolisme , Sarcoplasmic Reticulum Calcium-Transporting ATPases/métabolisme , Calcium/métabolisme
4.
Am J Physiol Heart Circ Physiol ; 320(4): H1321-H1336, 2021 04 01.
Article de Anglais | MEDLINE | ID: mdl-33481702

RÉSUMÉ

Arsenic exposure though drinking water is widespread and well associated with adverse cardiovascular outcomes, yet the pathophysiological mechanisms by which iAS induces these effects are largely unknown. Recently, an epidemiological study in an American population with a low burden of cardiovascular risk factors found that iAS exposure was associated with altered left ventricular geometry. Considering the possibility that iAS directly induces cardiac remodeling independently of hypertension, we investigated the impact of an environmentally relevant iAS exposure on the structure and function of male and female hearts. Adult male and female C56BL/6J mice were exposed to 615 µg/L iAS for 8 wk. Males exhibited increased systolic blood pressure via tail cuff photoplethysmography, left ventricular wall thickening via transthoracic echocardiography, and increased plasma atrial natriuretic peptide via enzyme immunoassay. RT-qPCR revealed increased myocardial RNA transcripts of Acta1, Myh7, and Nppa and decreased Myh6, providing evidence of pathological hypertrophy in the male heart. Similar changes were not detected in females, and nitric oxide-dependent mechanisms of cardioprotection in the heart appeared to remain intact. Further investigation found that Rcan1 was upregulated in male hearts and that iAS activated NFAT in HEK-293 cells via luciferase assay. Interestingly, iAS induced similar hypertrophic gene expression changes in neonatal rat ventricular myocytes, which were blocked by calcineurin inhibition, suggesting that iAS may induce pathological cardiac hypertrophy in part by targeting the calcineurin-NFAT pathway. As such, these results highlight iAS exposure as an independent cardiovascular risk factor and provide biological impetus for its removal from human consumption.NEW & NOTEWORTHY This investigation provides the first mechanistic link between an environmentally relevant dose of inorganic arsenic (iAS) and pathological hypertrophy in the heart. By demonstrating that iAS exposure may cause pathological cardiac hypertrophy not only by increasing systolic blood pressure but also by potentially activating calcineurin-nuclear factor of activated T cells and inducing fetal gene expression, these results provide novel mechanistic insight into the theat of iAS exposure to the heart, which is necessary to identify targets for medical and public health intervention.


Sujet(s)
Arsénites/toxicité , Hypertrophie ventriculaire gauche/induit chimiquement , Myocytes cardiaques/effets des médicaments et des substances chimiques , Composés du sodium/toxicité , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Polluants chimiques de l'eau/toxicité , Animaux , Calcineurine/métabolisme , Femelle , Régulation de l'expression des gènes , Cellules HEK293 , Humains , Hypertrophie ventriculaire gauche/métabolisme , Hypertrophie ventriculaire gauche/anatomopathologie , Hypertrophie ventriculaire gauche/physiopathologie , Préparation de coeur isolé , Mâle , Souris de lignée C57BL , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Facteurs de transcription NFATC/métabolisme , Facteurs sexuels , Transduction du signal , Facteurs temps
5.
J Mol Cell Cardiol ; 136: 95-101, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-31536744

RÉSUMÉ

TRIM72 is a membrane repair protein that protects against ischemia reperfusion (I/R) injury. We previously identified Cys144 (C144) on TRIM72 as a site of S-nitrosylation. To study the importance of C144, we generated a knock-in mouse with C144 mutated to a serine (TRIM72 C144S). We subjected ex vivo perfused mouse hearts to 20 min of ischemia followed by 90 min of reperfusion and observed less injury in TRIM72 C144S compared to WT hearts. Infarct size was smaller (54 vs 27% infarct size) and cardiac functional recovery (37 vs 62% RPP) was higher for the TRIM72 C144S mouse hearts. We also demonstrated that TRIM72 C144S hearts were protected against I/R injury using an in vivo LAD occlusion model. As TRIM72 has been reported to be released from muscle we tested whether C144 is involved in TRIM72 release. After I/R there was significantly less TRIM72 in the perfusate normalized to total released protein from the TRIM72 C144S compared to WT hearts, suggesting that C144 of TRIM72 regulates myocardial TRIM72 release during I/R injury. In addition to TRIM72's protective role in I/R injury, TRIM72 has also been implicated in cardiac hypertrophy and insulin resistance, and secreted TRIM72 has recently been shown to impair insulin sensitivity. However, insulin sensitivity (measured by glucose and insulin tolerance) of TRIM72 C144S mice was not impaired. Further, whole body metabolism, as measured using metabolic cages, was not different in WT vs TRIM72 C144S mice and we did not observe enhanced cardiac hypertrophy in the TRIM72 C144S mice. In agreement, protein levels of the TRIM72 ubiquitination targets insulin receptor ß, IRS1, and focal adhesion kinase were similar between WT and TRIM72 C144S hearts. Overall, these data indicate that mutation of TRIM72 C144 is protective during I/R and reduces myocardial TRIM72 release without impairing insulin sensitivity or enhancing the development of hypertrophy.


Sujet(s)
Cystéine/génétique , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Mutation , Lésion de reperfusion myocardique/métabolisme , Myocarde/métabolisme , Angiotensine-II/pharmacologie , Animaux , Cardiomégalie/génétique , Maladie des artères coronaires , Modèles animaux de maladie humaine , Techniques de knock-in de gènes , Insulinorésistance/génétique , Souris de lignée C57BL , Souches mutantes de souris , Lésion de reperfusion myocardique/génétique , Lésion de reperfusion myocardique/anatomopathologie , Myocarde/anatomopathologie
6.
Am J Physiol Heart Circ Physiol ; 316(5): H1053-H1064, 2019 05 01.
Article de Anglais | MEDLINE | ID: mdl-30822117

RÉSUMÉ

Arsenic is a common contaminant in drinking water throughout the world, and recent studies support a link between inorganic arsenic (iAS) exposure and ischemic heart disease in men and women. Female hearts exhibit an estrogen-dependent reduction in susceptibility to myocardial ischemic injury compared with males, and as such, female hearts may be more susceptible to the endocrine-disrupting effects of iAS exposure. However, iAS exposure and susceptibility to ischemic heart injury have not been examined in mechanistic studies. Male and female mice (8 wk) were exposed to environmentally relevant concentrations of sodium arsenite (0, 10, 100, and 1,000 parts/billion) via drinking water for 4 wk. Pre- and postexposure echocardiography was performed, and postexposure plasma was collected for 17ß-estradiol measurement. Hearts were excised and subjected to ischemia-reperfusion (I/R) injury via Langendorff perfusion. Exposure to 1,000 parts/billion iAS led to sex-disparate effects, such that I/R injury was exacerbated in female hearts but unexpectedly attenuated in males. Assessment of echocardiographic parameters revealed statistically significant structural remodeling in iAS-treated female hearts with no change in function; males showed no change. Plasma 17ß-estradiol levels were not significantly altered by iAS in male or female mice versus nontreated controls. Although total eNOS protein levels did not change in whole heart homogenates from iAS-treated male or female mice, eNOS phosphorylation (Ser1177) was significantly elevated in iAS-treated male hearts. These results suggest that iAS exposure can induce sex-disparate effects and modulate susceptibility to ischemic heart injury by targeting distinct sex-dependent pathways. NEW & NOTEWORTHY This is the first mechanistic study examining iAS exposure on myocardial ischemia-reperfusion injury in male and female mice. Following iAS exposure, ischemia-reperfusion injury was exacerbated in female hearts but attenuated in males. iAS treatment induced statistically significant cardiac remodeling in females, with no change in males. iAS treatment also enhanced phosphorylated eNOS levels at Ser1177, but only in male hearts. These results suggest that iAS alters susceptibility to myocardial I/R injury through distinct sex-dependent pathways.


Sujet(s)
Arsénites/toxicité , Lésion de reperfusion myocardique/induit chimiquement , Lésion de reperfusion myocardique/prévention et contrôle , Myocarde/anatomopathologie , Composés du sodium/toxicité , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Animaux , Cardiotoxicité , Modèles animaux de maladie humaine , Oestradiol/sang , Femelle , Préparation de coeur isolé , Mâle , Souris de lignée C57BL , Lésion de reperfusion myocardique/sang , Lésion de reperfusion myocardique/anatomopathologie , Myocarde/métabolisme , Monoxyde d'azote/métabolisme , Nitric oxide synthase type III/métabolisme , Phosphorylation , Facteurs sexuels
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...